Download as pdf or txt
Download as pdf or txt
You are on page 1of 19

Received: 18 July 2019 | Accepted: 15 August 2019

DOI: 10.1002/brb3.1408

REVIEW

The role of the brain–gut–microbiota axis in psychology: The


importance of considering gut microbiota in the development,
perpetuation, and treatment of psychological disorders

Michael Ganci1 | Emra Suleyman1 | Henry Butt2,3 | Michelle Ball1

1
Psychology Department, Institute for
Health and Sport, Victoria University, Abstract
Melbourne, Vic., Australia Introduction: The prevalence of psychological disorders remains stable despite
2
Bioscreen Yarraville (Aust) Pty Ltd,
steady increases in pharmacological treatments suggesting the need for auxiliary
Melbourne, Vic., Australia
3
Melbourne University, Melbourne, Vic.,
treatment options. Consideration of the brain–gut–microbiota axis (BGMA) has made
Australia inroads into reconceptualizing psychological illness from a more holistic perspective.

Correspondence
While our understanding of the precise role of gut microbiota (GM) in psychological
Michael Ganci, Psychology Department, illness is in its infancy, it represents an attractive target for novel interventions.
Institute for Health and Sport, Victoria
University, Melbourne, PO Box 14428,
Method: An extensive review of relevant literature was undertaken.
Melbourne, Vic. 8001, Australia. Results: Gut microbiota are proposed to directly and indirectly influence mood, cog‐
Email: Michael.ganci1@live.vu.edu.au
nition, and behavior which are key components of mental health. This paper outlines
Funding information
how GM may be implicated in psychological disorders from etiology through to treat‐
This paper did not receive any specific
grant from funding agencies in the public, ment and prevention using the Four P model of case formulation.
commercial, or not‐for‐profit sectors.
Conclusion: Moving forward, integration of GM into the conceptualization and treat‐
However, the first author was supported
by a postgraduate scholarship with no ment of psychological illness will require the discipline of psychology to undergo a
restrictions on publications cofunded by
significant paradigm shift. While the importance of the GM in psychological well‐
Bioscreen, an industry partner of Victoria
University. being must be respected, it is not proposed to be a panacea, but instead, an additional
arm to a multidisciplinary approach to treatment and prevention.

KEYWORDS
allostatic load, gut microbiota, precipitating factors, predisposing factors, protective factors,
psychology

1 | I NTRO D U C TI O N to the intertwined coevolution between humans and our resident


microbes. It is suggested that the sharp increase in various disease
Burgeoning research regarding the role of the gut and its microbial states over the last 50–100 years (Campell, 2014; Linneberg et al.,
inhabitants in the pathophysiology of psychological illness is gain‐ 2000) can be, at least in part, explained by relatively recent dietary
ing momentum. Early evidence points to gut microbiota (GM) as a and lifestyle changes in the context of human evolution (Broussard
possible missing link in the conceptualization and treatment of psy‐ & Devkota, 2016). Currently, humans, particularly those in indus‐
chological illness that sees disparities between conventional treat‐ trialized countries, are living in an environment to which they have
ment methods and prevalence rates. When discussing the role of not adaptively evolved (Gluckman, Low, Buklijas, Hanson, & Beedle,
GM in behavior, health, and disease, it is important to pay respect 2011). An unintended consequence of industrialization, these

This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium,
provided the original work is properly cited.
© 2019 The Authors. Brain and Behavior published by Wiley Periodicals, Inc.

Brain and Behavior. 2019;9:e01408.  wileyonlinelibrary.com/journal/brb3 | 1 of 19


https://doi.org/10.1002/brb3.1408
2 of 19 | GANCI et al.

changes are putting the GM under evolutionary pressure to shift disorders such as IBS and psychological conditions (Garakani et al.,
from a previously mutualistic relationship with their human host to 2003; Lee et al., 2015).
a more antagonistic one (Broussard & Devkota, 2016; Quercia et al., Within the field of psychology, an integral part of intervention is
2014). This is due to human evolution requiring significantly more case formulation. Essentially, case formulation involves the synthe‐
time (Uyeda, Hansen, Arnold, & Pienaar, 2011) compared to single‐ sis of information about a patient (typically gained through clinical
celled organisms such as GM that evolve and adapt to environmental interviews and formalized cognitive testing) in a meaningful way to
and internal states much more rapidly (within as little as 24 hr; David facilitate the development of a treatment plan. A commonly used
et al., 2014; Wu et al., 2011). These variations in the evolutionary model in structuring a case formulation is that of the Four Ps which
pressures and capabilities of the two components (host and GM) of a represent the predisposing, precipitating, perpetuating, and pro‐
single ecosystem (the holobiont; Theis et al., 2016) result in systemic tective factors related to a client's presenting problem and thus the
disharmony. targets of psychological intervention. As information is drawn from
This systemic disharmony leads to symptomatology and disease clinical interviews and cognitive testing, the functioning of a client's
states that are the primary target for intervention in current medi‐ gut is seldom considered in their formulation; thus, important diag‐
cal models, which precludes effective etiology‐focused prevention nostic and treatment options may be missed. It is the contention of
(Marvasti & Stafford, 2012). This is exacerbated given that current this paper to demonstrate that GM are intimately linked with each of
medical models are heavily skewed toward treatment over preven‐ these four pillars of psychological intervention and thus each stage
tion (Singh, 2010). While there is disagreement over whether the of disease, from etiology through to treatment and prevention. This
occurrence of common mental disorders are increasing or whether paper adds to the burgeoning research into the brain–gut–microbi‐
their prevalence remains consistent (Friedrich, 2017; Harvey et al., ota axis (BGMA; Kelly, Clarke, Cryan, & Dinan, 2016) demonstrat‐
2017), the consumption of antidepressant drugs doubled in most, if ing that the discipline of psychology is on the cusp of a significant
not all, Organisation for Economic Co‐operation and Development paradigm shift, moving away from CNS‐centric approaches toward
(OECD) countries between 2000 and 2015 (OECD, 2017). Despite a more holistic conceptualization of health and disease which inte‐
this, depression has recently taken over as the leading cause of dis‐ grates other body systems. We echo the sentiment of Allen, Dinan,
ability worldwide with anxiety also in the top 10 leading causes of Clarke, and Cryan (2017) who call for a challenge to the reductionist
disability (World Health Organisation (WHO) 2017). Furthermore, approaches in psychology in favor of a multidisciplinary approach to
subclinical psychological symptoms that do not meet the full conceptualizing and treating psychological disorders. In taking the
Diagnostic and Statistical Manual for Mental Disorders (DSM) or unique approach of the Four P model of case formulation, this paper
International Classification of Diseases (ICD) diagnostic criteria are intends to review existing research on associations between GM and
also prevalent (Angst, Merikangas, & Preisig, 1997; Haller, Cramer, psychological outcomes, compiling it in a way that is more accessible
Lauche, Gass, & Dobos, 2014; Mathieson, Collings, & Dowell, 2009). to psychologists, especially those who have little previous knowl‐
These subclinical symptoms lead to impairment in psychosocial and edge regarding the BGMA.
work functioning, are a major determinant of sick leave, contribute
to the use of psychotropic drugs and primary health care services,
1.1 | The brain–gut–microbiota axis
and reduce quality of life (Haller et al., 2014; Johansen & Dittrich,
2013; Mendlowicz & Stein, 2000). The BGMA is increasingly being recognized as playing an important
The conceptualization of the human body as being made up of role in homeostasis and consequentially, health and disease states
separate and distinguishable systems is likely to have contributed (e.g., Mu, Yang, & Zhu, 2016; Rea, Dinan, & Cryan, 2016). The BGMA
to our current poor appreciation of the complexity of mechanisms refers to the relationship between the brain and the gut while ac‐
that underlie the etiology and progression of disease. Psychology, knowledging the important moderating role of GM (e.g., Carabotti,
just like many other healthcare professions and medical science dis‐ Scirocco, Maselli, & Severi, 2015; Grenham, Clarke, Cryan, & Dinan,
ciplines, specialize and focus on specific body systems. For example, 2011). Largely recognized as a bidirectional relationship (e.g., Rhee,
treatment of psychological symptoms and disorders tends to be cen‐ Pothoulakis, & Mayer, 2009), research continues to uncover the true
tral nervous system (CNS)‐centric with the brain being the principle complexities of this communication network which Rea et al. (2016)
target for both psychotherapy and psychopharmacology while pe‐ more accurately define as a multidirectional relationship. It is multi‐
ripheral systems, such as the gut, receive little attention. While there directional in the sense that each component of this extensive com‐
is no doubt that specialization has its benefits and has contributed munication network has the ability to moderate and manipulate the
to the progression of medical knowledge, it also has its drawbacks. function of the other systems involved. Communication between
Operating within these constrictive distinctions imposed by disci‐ the brain and the gut is maintained via a complex network including
plinary specialization means that the complex interplay between the CNS, autonomic nervous system (ANS), enteric nervous system
various body systems essential to proper functioning is overlooked. (ENS), hypothalamic–pituitary–adrenal (HPA) axis, neural, endocrine
The true etiology of disease is likely to come from dysregulations in and immune systems (e.g., Carabotti et al., 2015; Cryan & Dinan,
this interplay which may also go some way to explaining high lev‐ 2012; Mayer, 2011; Moloney, Desbonnet, Clarke, Dinan, & Cryan,
els of comorbidity, particularly between functional gastrointestinal 2014). Essentially, the BGMA provides a network for signals from
GANCI et al. | 3 of 19

the brain to influence the motor, sensory, and secretory functions diversity has been further reduced over the past 50 years with an
of the gut while simultaneously allowing signals and metabolites ever increasing preference for convenience and taste (Glanz, Basil,
from the GM to influence brain development, biochemistry, func‐ Maibach, Goldberg, & Snyder, 1998; Heiman & Greenway, 2016;
tion, and behavior (e.g., Cryan & O'Mahony, 2011; Grenham et al., Poti, Mendez, Ng, & Popkin, 2015). Essentially, this means that
2011; Marques et al., 2014). This communication system presents an current human diets are not providing GM with the resources they
exciting and novel target for psychological intervention, providing a require to perform their myriad of complex tasks involved in host
deeper understanding of the biological underpinnings of psychologi‐ homeostasis and consequently health and disease. Adding support
cal illnesses. It is hoped that developing a greater understanding of to this contention, Jacka et al. (2017) conducted a clinical trial which
the BGMA as it relates to the Four Ps of case formulation will provide demonstrated that adherence to a modified Mediterranean diet
psychologists with an additional tool in the treatment of their clients. resulted in significantly greater improvement in depression ratings
from baseline compared to a social support control group.
While diet is one way through which a host can modulate their
1.2 | Is diet the chicken, or the egg?
GM, microbes are themselves able to influence eating behaviors of
The relationship between diet and GM is an intriguing one, ironically their host (Alcock et al., 2014). Microbes in the gut must cooper‐
reminiscent of the idiom regarding which came first, the chicken or ate and share limited resources (space and nutrients) to promote
the egg. Once thought to be unidirectional (diet influencing the com‐ stable coexistence and ecological diversity (Allen & Nowak, 2013).
position of the microbiota), recent evidence suggests the relation‐ This means that these microorganisms are under selective pres‐
ship could in fact be bidirectional, with microbes also being able to sure to ensure their own survival and must therefore compete for
influence food choice and dietary‐related behaviors (Alcock, Maley, available resources (Hibbing, Fuqua, Parsek, & Peterson, 2010). As
& Aktipis, 2014). such, microbes are proposed to manipulate the eating behavior of
Diet is arguably one of the most important environmental fac‐ the host by either generating cravings for foods that they thrive on
tors in shaping the composition and metabolic activities of GM (De or those which suppress their competitors, or by influencing mood
Filippo et al., 2017; Garcia‐Mantrana, Selma‐Royo, Alcantara, & which leads to the intake of foods that enhance that species' fitness
Collado, 2018; Voreades, Kozil, & Weir, 2014). As such, diet must (Alcock et al., 2014; Leitao‐Goncalves et al., 2017).
be taken into consideration when discussing potential interventions
involving GM. An extensive review of the relationship between diet
1.2.1 | Dietary‐derived short‐chain fatty acids
and GM is beyond the scope of this paper, but can be found else‐
where (e.g., Sheflin, Melby, Carbonero, & Weir, 2017; Singh et al., A continued loss of fiber from the Western diet will inevitably lead
2017; Wu et al., 2011). Here, diet is discussed insofar as to high‐ to continued depletion of short‐chain fatty acids (SCFAs; Broussard
light to psychologists the importance of this environmental factor & Devkota, 2016) with downstream effects on the development and
in the formulation and treatment of their client's presenting prob‐ perpetuation of psychological illnesses through their immunoregula‐
lem. While it is not being suggested that psychologists become well tory effects (Rogers et al., 2016). SCFAs (acetic, butyric, and pro‐
versed in dietetics, gathering general information on a client's diet pionic acids in particular) are one of the main metabolites of GM
may provide further insight into the development and perpetuation (Carabotti et al., 2015; Smith et al., 2013). They are the end products
of their presenting problem, and presents as an additional arm to a of dietary fiber fermentation and have been shown to have many
multidisciplinary and holistic treatment approach. beneficial effects on host health (Bourassa, Alim, Bultman, & Ratan,
Both the content and diversity of an individual's diet are be‐ 2016; den Besten et al., 2013).
lieved to be important in maintaining a well‐balanced GM (Heiman & In the brain, SCFAs demonstrate neuroprotective properties
Greenway, 2016; Oriach, Robertson, Stanton, Cryan, & Dinan, 2016). (Sun et al., 2015) with butyrate in particular having a protective ef‐
Diet quality has also been highlighted as a potential risk or protective fect on psychological and neurodegenerative disorders (Bourassa
factor for conditions such as depression (Jacka et al., 2017; Koopman et al., 2016). Peripherally, SCFAs are believed to influence the
& El Aidy, 2017; Lai et al., 2014). In regards to dietary content, the size and function of regulatory T cells which play a crucial role
industrial revolution saw a significant increase in highly processed in regulating inflammation and immune homeostasis (Hakansson
cereals rich in carbohydrates, refined sugars, sodium, omega‐6, and & Molin, 2011; Smith et al., 2013). Additionally, SCFAs (together
trans‐fatty acids. Concurrently, potassium, complex carbohydrates, with enzymes also produced by the GM) enhance intestinal bar‐
fiber, omega‐3, and unsaturated fatty acids were considerably re‐ rier functioning through their regulation of tight junction (TJ) pro‐
duced (Rubio‐Ruiz, Peredo‐Escárcega, Cano‐Martínez, & Guarner‐ teins (e.g., Anderson et al., 2010; Bischoff et al., 2014; Peng, Li,
Lans, 2015). These changes are reflective of what is today termed Green, Holzman, & Lin, 2009). Abnormal intestinal permeability
the “Western‐style diet,” one that has been shown to impair immune (leaky gut) results in increased translocation of toxins and GM
function and promote inflammation (Myles, 2014). This is concerning across the epithelial barrier which consequently trigger an inflam‐
given that inflammation is believed to underlie and perpetuate many, matory immune response that can dysregulate ENS and systemic
if not all, psychological and neurodegenerative illnesses (Almond, immune functioning (Berkes, Viswanathan, Savkovic, & Hecht,
2013; Miller & Raison, 2016; Rea et al., 2016). Worryingly, dietary 2003; Carabotti et al., 2015; Fasano, 2012; Smith et al., 2013).
4 of 19 | GANCI et al.

This immune response is believed to be the instigator of resultant A recently proposed way in which GM may manipulate host
symptom expression including psychological disorders such as de‐ behavior is via their relationship with personality traits. Kim et al.
pression (e.g., Maes et al., 2013; Mulak & Bonaz, 2015; Sheedy et (2018) found an increased abundance of Gammaproteobacteria in
al., 2009). those with high neuroticism, as well as those with low extraversion.
Also via their influence on TJ proteins, SCFAs are believed to reg‐ Low conscientiousness was associated with an increased abundance
ulate the permeability of the blood–brain barrier (BBB; Braniste et of Proteobacteria and a decreased abundance of Lachnospiraceae
al., 2014). Dysregulation of the BBB has been associated with neu‐ while those with high levels of openness demonstrated greater phy‐
ropsychological conditions including Alzheimer's disease (Kuhnke logenic diversity and richness (Kim et al., 2018). As this was the first
et al., 2007) and autism (Fiorentino et al., 2016). Schoknecht and study to have investigated the link between GM and personality di‐
Shalev (2012) suggest that depression and schizophrenia may also rectly, further research is required to elucidate these relationships.
be related to BBB dysfunction. Although further research is needed, The relationship between personality and GM presents as an intrigu‐
these associations are highly plausible given the BBB is responsi‐ ing area of exploration, given that personality traits have a strong
ble for regulating access of circulating macromolecules and poten‐ association with behavioral patterns in addition to physiological
tial neurotoxins to the brain (Fiorentino et al., 2016; Patel & Frey, and psychological health outcomes (e.g., Ferguson, 2013; Kim et al.,
2015). As evidence of microbial involvement, Braniste et al. (2014) 2018; Srivastava & Das, 2015).
found that germ‐free (GF) mice (those devoid of bacterial coloni‐ Additional evidence linking GM composition and behavior comes
zation and therefore lacking conventional gut flora) have increased from the study of patients following gastric bypass surgery. Behavioral
BBB permeability compared to specific pathogen‐free (SPF) mice changes following gastric bypass surgery include patients feeling less
that have conventional GM colonization free of any known patho‐ hungry and having a preference for healthier foods (Behary & Miras,
gens. Colonization of GF mice with known SCFA‐producing bacterial 2015) which is likely related to changes in neural responses to food
strains (Clostridium tyrobutyricum and Bacteroides thetaiotaomicron) (particularly high‐calorie foods) in key areas of the mesolimbic re‐
was found to normalize BBB function (Braniste et al., 2014). There ward pathway (Ochner et al., 2011, 2012). It is tempting to speculate
is also evidence to suggest that SCFAs are involved in glucose me‐ that these changes are associated with the compositional changes
tabolism, reducing adiposity, appetite regulation, and energy ho‐ in GM following gastric bypass surgery (Furet et al., 2010; Liou et
meostasis (Byrne, Chambers, Morrison, & Frost, 2015; Chambers et al., 2013; Zhang et al., 2009). Additionally, improvements in qual‐
al., 2015; Kondo, Kishi, Fushimi, Ugajin, & Kaga, 2009; Morrison & ity of life and levels of depression have been shown to persist two
Preston, 2016). years after surgery (Karlsson, Sjostrom, & Sullivan, 1998; Mokhber,
Shaghayegh, Talebi, & Tavassoli, 2016). These improvements may be
the result of reduced adipose tissue which has downstream effects
1.3 | Behavior
on GM and their role in inflammation and other related functions.
Studies using GF mice have provided the greatest depth of infor‐ While causational evidence is currently unavailable, correlational re‐
mation regarding the influence of GM on host behavior. Such pre‐ search linking GM and mood (Jiang et al., 2015) suggest that changes
clinical work gives useful insights into the physiological mechanisms in GM composition following gastric bypass surgery may also have a
through which the BGMA functions. For example, GF mice dem‐ direct influence on mood. Improvements in mood may consequently
onstrate altered expression of brain‐derived neurotrophic factor encourage healthier food choices and eating behaviors (Christensen
(BDNF) and SCFA while also exhibiting altered HPA axis functioning, & Brooks, 2006), exemplifying the potential for a cyclical relationship
anxious and depressive behaviors, and social functioning (Arentsen, involving diet, GM, and mood that is beneficial to overall health.
Raith, Qian, Forssberg, & Diaz Heijtz, 2015; Luczynski et al., 2016; Given the possible role of GM in eating behaviors, and their
Neufeld, Kang, Bienenstock, & Foster, 2011; Sudo et al., 2004). ability to influence hunger and satiety (Cani et al., 2009) and neuro‐
In both animal and human models, manipulation of GM has also peptide and endocrine regulation (Holzer & Farzi, 2014), a relatively
been demonstrated to alter levels of stress hormones corticotro‐ new line of inquiry has emerged investigating GM involvement in
pin‐releasing factor (CRF) and cortisol (Yarandi, Peterson, Treisman, eating disorders which are traditionally recognized as psychological
Moran, & Pasricha, 2016). Many of these abnormalities have been disorders (Kleiman et al., 2015; Lam, Maguire, Palacios, & Caterson,
shown to be rectified by colonization with the feces from SPF mice 2017). Associations between GM composition and eating disorder
or with specific probiotics (e.g., Bercik et al., 2011; Desbonnet et al., psychopathology were also found by Kleiman et al. (2015), further
2010; Sudo et al., 2004). However, Bravo et al. (2011) found that suggesting that the GM play a role in the psychology of food choice
ingestion of probiotics was only beneficial in mice which had an in‐ and eating behaviors. Given that diet is a key determinant of GM
tact vagus nerve, demonstrating the importance of the vagal path‐ composition and that eating disorders are categorized by extreme
way in brain–gut communication. Additionally, Sudo et al. (2004) dietary changes, the GM present as a logical target for inclusion in
demonstrated that recolonization was only effective if it occurred multifaceted intervention.
within a critical period, providing evidence for a fundamental role While this paper focuses mainly on unconscious mechanisms un‐
of GM in the development of crucial systems involved in behavioral derlying the relationship between GM and psychological outcomes
outcomes. (such as interoceptive processes and neurotransmitter production), it
GANCI et al. | 5 of 19

Endocrinological Neuronal Immunological

Neurotransmi er
produ on Altered func on
HPA axis Inflamma on
(Allosta response) (Allosta response)
Blood brain
barrier

Homeosta
emo ons Allosta c
overload

Microbial Altered
Interoc on Leaky gut
SCFA
dysbiosis
produ on

Symptom
expression

F I G U R E 1 Factors influencing the multidirectional communication between the brain and the gut. Double‐headed arrows demonstrate
a bidirectional relationship, with broken arrows demonstrating proposed but not yet established relationships. The figure demonstrates
the three main well‐established pathways of communication between the brain and the gut, being endocrinological, neuronal, and
immunological. The figure also illustrates the bidirectional relationships between microbial dysbiosis and the HPA axis, neurotransmitter
production, the function of the blood–brain barrier, and inflammation which are believed to alter their functioning as an allostatic response
to homeostatic emotions. It is proposed that microbial dysbiosis itself is able to be detected via the interoceptive system which then triggers
these homeostatic emotions

is acknowledged that conscious mechanisms also have potential psy‐ action of several key neurotransmitters (e.g., Anderson & Maes,
chological implications. For example, gastrointestinal symptoms can 2015; Lyte, 2011; O'Mahony, Clarke, Borre, Dinan, & Cryan, 2015).
be noticeably unpleasant and, particularly in IBS sufferers, can lead to GM regulate the metabolism and concentration of amino acids which
impairment in daily functioning (Ballou, Bedell, & Keefer, 2015), anxi‐ serve as precursors for several neurotransmitters including gamma‐
ety and depression (Roohafza et al., 2016), avoidance behaviors (Van aminobutyric acid (GABA), serotonin, melatonin, and dopamine,
Oudenhove et al., 2016), and poor quality of life (Canavan, West, & among others (Clarke et al., 2014; Evrensel & Ceylan, 2015; Jenkins,
Card, 2015). Conscious mechanisms can also lead to positive psycho‐ Nguyen, Polglaze, & Bertrand, 2016; Zagajewski et al., 2012). As
logical outcomes as exemplified by patients following gastric bypass such, it is highly plausible that GM are able to influence brain chemis‐
surgery. For example, noticeable changes in body composition can try, which consequentially regulates cognition, mood, and behavior.
result in more positive body image which in itself is related to psycho‐ As depicted in Figure 1, demonstrating bidirectionality, GM can also
logical well‐being, particularly after body contouring surgery (Jumbe, be directly affected by neurochemicals which alter bacterial growth
Hamlet, & Meyrick, 2017; Sarwer & Steffen, 2015; Song et al., 2016). and pathogenicity (Lyte, 2011). Table 1 displays some of the key neu‐
These changes can then encourage long‐term weight loss maintenance rotransmitters synthesized by GM whose dysregulation is associated
behavior (Palmeira et al., 2010). with psychological disorders. While neurotransmitters produced in
the gut may not directly influence brain chemistry as they do not
pass through the BBB, they are able to influence the CNS through
1.4 | Neurotransmitters
mechanisms including direct stimulation of the vagus nerve, as well
Perhaps the most obvious association between GM and psychologi‐ as using indirect circulatory and immune pathways (Sampson &
cal illnesses is the ability of GM to manipulate the production and Mazmanian, 2015). For example, tryptophan, the precursor molecule
6 of 19

TA B L E 1 Gut bacteria associated with the synthesis of key neurotransmitters


|

Neurotransmitter Psychiatric conditions related


Genus/species (precursor) CNS effect Peripheral effect to dysregulation References

Candida; Streptococcus; Serotonin Motor control, cerebellar Circadian rhythm, gut motility, Depression, IBS, autism, Arreola et al., (2015), Gulesserian,
Escherichia; Enterococcus; (tryptophan) regulation, synaptogen‐ body temperature, visceral Down's syndrome Engidawork, Cairns, and Lubec (2000),
Lactobacillus bulgaricus esis, addiction, emotion, pain, appetite, modulation of Halford and Blundell, (2000), Hood
memory, stress immune response et al. (2006), Jenkins et al. (2016),
Leonard, (2010), Mazzoli and Pessione
(2016), Marks et al. (2009), Meneses
and Liy‐Salmeron (2012), Müller and
Homberg (2015), Rogers et al. (2016),
Stasi, Rosselli, Zignego, Laffi, and Milani
(2014), Warren and Singh, (1996),
Whitaker‐Azmitia (2001)
Corynebacterium glu‐ l‐glutamate Excitatory, brain develop‐ Generalized anxiety disorder, Abdou et al. (2006), Boonstra et al.
tamicum; Lactobacillus ment, synaptic plasticity depression, bipolar, schizo‐ (2015), Cherlyn et al. (2010), Femenía,
plantarum; Lactobacillus phrenia, neurodegeneration Gómez‐Galán, Lindskog, and Magara
paracasei; Lactobacillus lactis; (2012), Hyland and Cryan (2010),
Brevibacterium lactofermentum; Meldrum (2000), Yoto et al. (2012)
Brevibacterium flavum
Lactobacillus; Bifidobacterium; GABA Inhibitory, anxiolytic Myorelaxant, moderates intes‐
Escherichia coli; Pseudomonas (l‐glutamate) tinal motility, gastric empty‐
ing, gastric acid secretion, and
inhibits GI carcinogens and
tumor growth
Bacillus, Serratia, E. coli Dopamine (l‐Dopa) Reward‐motivated behavior, Stimulates exocrine secre‐ Schizophrenia, Parkinson's Di Chiara and Bassareo (2007),
motor behavior, cognition, tion, inhibits gut motility, and disease, depression, anxiety, Eisenhofer et al. (1997), Freestone
emotion modulates sodium absorption addiction (2013), Grace (2016), Lyte (2011), Meyer
and mucosal blood flow and Feldon (2009), Scheperjans et al.
(2015), Shishov, Kirovskaya, Kudrin, and
Oleskin (2009)
Bacillus; E. coli; Saccharomyces Norepinephrine Stress hormone, attentive‐ Mediates growth and virulence Depression, schizophrenia Freestone (2013), Moret and Briley
(dopamine) ness, emotion, sleep, of potentially pathogenic (2011), Yamamoto and Hornykiewicz
learning bacteria (2004)
(dependent on tryptophan Melatonin Circadian rhythm, mood Gastrointestinal function, pro‐ IBS, multiple sclerosis, autism, Fornaro, Prestia, Colicchio, and Perugi
production and serotonin (serotonin) tects against gut permeability, Alzheimer's, mood disorders (2010), Ghorbani, Salari, Shaygannejad,
synthesis) anti‐inflammatory, antioxi‐ and Norouzi (2013), Ortiz, Benítez‐King,
dant, analgesic Rosales‐Corral, Pacheco‐Moisés, and
Velázquez‐Brizuela (2008), Veatch,
Goldman, Adkins, and Malow (2015),
Wong, Yang, Song, Wong, and Ho (2015)
GANCI et al.
GANCI et al. | 7 of 19

to serotonin (which is itself the precursor to melatonin), is able to which ultimately leads to altered functioning of factors (e.g., inflam‐
pass through the BBB and as such it is likely that metabolites of GM mation) that mediate these pathways. These alterations are believed
directly influence brain chemistry (Sampson & Mazmanian, 2015). to be an allostatic response to a deviation from a “typical” microbi‐
ome, which over time, leads to symptom expression as a result of
allostatic overload. The bidirectional relationship between microbial
1.5 | Interoception and allostatic responses
dysbiosis and the factors which alter the three main communication
It is currently unknown whether the interoceptive system is able pathways between the gut and the brain illustrate the multidirec‐
to detect microbial dysbiosis (imbalances resulting from the under‐ tional nature of the BGMA.
or overabundance of certain microbial species; DeGruttola, Low,
Mizoguchi, & Mizoguchi, 2016), but considering that gut microbes
are an essential part of human physiology which moderate sev‐ 2 | G M TH RO U G H TH E LE N S O F A C A S E
eral homeostatic emotions (Craig, 2002; Mayer, Naliboff, & Craig, FO R M U L ATI O N FR A M E WO R K
2006; Noakes, 2012; Paulus & Stein, 2010) it is a strong possibility.
Homeostatic emotions are background emotions that may or may It is not the intention of this paper to propose that the BGMA must be
not enter conscious awareness but influence an individual's energy at the forefront of consideration for each and every client. Instead, it
levels, mood, and disposition (Mayer et al., 2006). Signals from in‐ is proposed that the relevance of the BGMA is determined on a case‐
ternal organs, particularly the gut, continuously communicate with by‐case basis. The role of the BGMA in a client's presenting prob‐
various regions of the brain including the limbic system, autonomic lem may be less relevant when there are clear social and emotional
and neuroendocrine centers in the hypothalamus, brainstem, and etiological factors, such as the presence of significant stressors for
cortex (Craig, 2002; Holzer & Farzi, 2014; Mayer & Tillisch, 2011). a client presenting with anxiety or depression. However, it is worth
It is plausible that through the process of interoception, GM are noting that stress can alter the composition of a person's GM (Bailey
able to influence human cognition, emotion, and mood through et al., 2011), which may or may not be clinically relevant, but should
their involvement in systemic functioning through their various me‐ be considered if comorbidities are present. It may also be less rel‐
tabolites (Holzer, 2017; Paulus & Stein, 2010). As such, increasing evant for clients who respond well to traditional psychological treat‐
rates of disease might be explained by the allostatic load hypoth‐ ments such as cognitive behavioral therapy. Alternatively, cases in
esis (McEwen, 1998). The allostatic load hypothesis proposes that which the role or the BGMA may be more pertinent are when social
rather than having a stable set point, body systems have a range and emotional etiological factors are less clear or absent, and also
of set points allowing them to actively adapt to environmental and for clients who do not respond well to conventional psychological
internal states. Allostatic load is a term used to refer to the cumu‐ approaches. In cases where a treating clinician considers investiga‐
lative cost of allostasis to the body (“wear and tear”; McEwen & tion of the GM to be appropriate, clients should be referred for stool
Wingfield, 2003). While adaptive in the short term, allostasis can sampling and analysis and an open dialogue established between the
become maladaptive, leading to disease, when allostatic measures clinician and the microbiologist performing the analysis. The follow‐
are required to vary widely and frequently, or are at extreme val‐ ing information serves to highlight possible associations between
ues for long periods of time (James, 2013). Additionally, allostatic GM and each of the Four Ps.
systems can become dysfunctional when they lose their ability to
change or regulate change (James, 2013). The result of either of
2.1 | Predisposing
these scenarios is allostatic overload which can lead to symptom
expression, as depicted in Figure 1 (Berger, Juster, & Sarnyai, 2015; As part of their first line of questioning, mental health professionals
McEwen, 2005; McEwen & Wingfield, 2003). The fact that research attempt to explore a client's family history of psychological illness
has failed to define the precise composition of a healthy GM due to establish whether that individual has an underlying genetic pre‐
to immense interindividual differences (Lloyd‐Price, Abu‐Ali, & disposition (vulnerability) to developing a psychological condition(s).
Huttenhower, 2016) suggests that the GM may in fact be the most Genetic predisposition to a multitude of psychological conditions has
allostatic system within the body. Microbial dysbiosis then could be been well established (Hyman, 2000). While research into the role
considered an extreme and prolonged shift away from what would of GM in genetic predisposition is scarce, several lines of evidence
be considered a relatively healthy composition which loses its abil‐ suggest that GM may play an integral part in a person's vulnerability
ity to regulate change in various other host systems and functions. to the development of psychological illnesses. Firstly, host genetics
It is perhaps this dysregulation that manifests itself in psychological have been demonstrated to influence the composition and meta‐
illness. bolic activities of GM (Goodrich et al., 2014; Ussar, Fujisaka, & Kahn,
Figure 1 depicts the three overarching pathways (endocrinolog‐ 2016) which have important consequences on host physiology, brain
ical, neuronal, and immunological) of bidirectional communication development, and health (e.g., Krishnan, Alden, & Lee, 2015; Sekirov,
between the brain and the gut, each of which is altered during a Russell, Antunes, & Finlay, 2010). However, the specific mechanisms
state of microbial dysbiosis. Although it remains unconfirmed, it is behind this relationship remain unclear (Dąbrowska & Witkiewicz,
proposed that microbial dysbiosis can be detected via interoception 2016).
8 of 19 | GANCI et al.

alterations in GM (Buford, 2017). Additionally, aging is associated


2.1.1 | Vertical transmission
with changes in the serotonergic system which is also believed to
Additionally, there is evidence to suggest that much like genetics are contribute to increased prevalence of psychological disorders in the
passed down from parents to offspring, GM are vertically transmit‐ elderly (O'Mahony et al., 2015). The serotonergic system is regulated
ted from mother to infant (e.g., Asnicar et al., 2017; Mueller, Bakacs, by GM (Table 1), therefore making it vulnerable to compositional and
Combellick, Grigoryan, & Dominguez‐Bello, 2015). The transmis‐ metabolic changes (O'Mahony et al., 2015; Rogers et al., 2016).
sion of microbiota from mother to infant during birth represents Given the sheer complexity of the systemic functioning of the
the most important point of microbial colonization in the infant gut, human body, there are likely to be several other processes through
which continues over the first three years of life (Yatsunenko et al., which GM may be involved in predisposing individuals to the de‐
2012). This critical establishment period of GM is in line with the velopment of psychological illnesses. Further research into how GM
critical development period of the human host (Rea et al., 2016). influence predisposition to psychological illness will be useful in in‐
It is during this time that GM play key roles in the development of forming preventative strategies to circumvent the growing burden
the CNS, HPA axis, and immune system (Borre et al., 2014; Cox et of such conditions. However, GM not only play a role in predisposing
al., 2014; Furusawa et al., 2013; Houghteling & Walker, 2015). As an individual to certain psychological illnesses as highlighted above,
such, aberrations in typical colonization of GM during this critical pe‐ but also in the onset and maintenance of those negative health
riod may also result in abnormal development of these key systems outcomes.
(Tamburini, Shen, Wu, & Clemente, 2016). Factors resulting in aber‐
rations to conventional microbial colonization of the gut during this
2.2 | Precipitating and perpetuating
period, such as birth by cesarean section and antibiotic treatment
during infancy, have been associated with increased rates of chronic While psychologists tend to focus heavily on social and environ‐
and atopic diseases (Kolokotroni et al., 2012; Sevelsted, Stokholm, mental factors involved in the onset and maintenance of psycho‐
Bonnelykke, & Bisgaard, 2015; Vangay, Ward, Gerber, & Knights, logical disorders, biological factors, such as GM composition, also
2015). A recent study by Polidano, Zhu, and Bornstein (2017) high‐ contribute to these stages of disease. The maintenance of a diverse
lights the microbiota as a potentially important factor in the negative microbial ecosystem in the gut is essential for optimal host func‐
relationship they found between cesarean birth and a range of cog‐ tion (Moloney et al., 2014; Sekirov et al., 2010). On the other hand,
nitive outcomes compared to those born vaginally. reduced diversity and microbial dysbiosis have been implicated in
There is a growing consensus that maternal GM may have long‐ various psychological, neurological, metabolic, functional gastroin‐
term health consequences for the child (Stanislawski et al., 2017). testinal disorders, and autoimmune disease states (Blumstein, Levy,
It is therefore reasonable to suggest that vertically transmitted GM Mayer, & Harte, 2014). These include, but are not limited to, IBS
may act as a mechanism for intergenerational predisposition to psy‐ (Jeffery et al., 2012; Tana et al., 2010), autism (Finegold et al., 2002),
chological disorders. Further research is required as it is difficult to schizophrenia (Castro‐Nallar et al., 2015), myalgic encephalomyeli‐
determine whether these intergenerational patterns are due to the tis/chronic fatigue syndrome (ME/CFS; Fremont, Coomans, Massart,
vertical transmission of GM or whether they are due to learned be‐ & Meirleir, 2013; Wallis, Butt, Ball, Lewis, & Bruck, 2016), multiple
haviors and lifestyle factors shared among family members. This is sclerosis (Jangi et al., 2016), dementia (Alkasir, Li, Li, Jin, & Zhu,
evidenced by same‐household members showing a higher similarity 2017), stress (Knowles, Nelson, & Palombo, 2008), anxiety (Burch,
of GM composition to those outside of the household (Abeles et al., 2016), depression (e.g., Jiang et al., 2015), obesity (Ley, Turnbaugh,
2016; Song et al., 2013; Yatsunenko et al., 2012). Klein, & Gordon, 2006), diabetes (Larsen et al., 2010), coronary ar‐
tery disease (Cui, Zhao, Hu, Zhang, & Hua, 2017; Emoto et al., 2017),
and cancer (particularly colorectal cancer; Gagniere et al., 2016;
2.1.2 | Aging
Garrett, 2015). Current findings linking GM and disorders are, at
Aging, in and of itself, can also predispose an individual to several this stage, associative and causative links are yet to be established.
psychological illnesses. For example, neurodegenerative illnesses, Additionally, it is unknown whether alterations in GM composition
such as Alzheimer's disease, are considered an evolutionary acci‐ are the cause or consequence of disease. Given available evidence
dent occurring as a result of increased longevity (Giunta et al., 2008; however, it is likely that this is a bidirectional, and cyclical, relation‐
Gluckman et al., 2011; Niccoli & Partridge, 2012). There is evidence ship as depicted in Figure 1.
to suggest that psychological disorders are also more prevalent in
the elderly (e.g., Andreas et al., 2017). A contributing factor toward
2.2.1 | Stress
the increased prevalence of disease in the elderly is that the nor‐
mal aging process is associated with compositional changes and re‐ Stress has long been recognized as both a precipitating and per‐
duced diversity of GM (Biagi et al., 2010). In parallel, normal aging petuating factor to various psychological conditions (Anisman &
is characterized by chronic low‐grade inflammation, a phenomenon Zacharko, 1982; Corcoran et al., 2003). Given the crucial role of GM
commonly referred to as “inflammaging” (Franceschi et al., 2007). It in the development and functioning of the HPA axis (Sudo, 2012),
is believed that this inflammation is, at least in part, attributable to as well as their modulation of stress hormones CRF and cortisol
GANCI et al. | 9 of 19

(Carabotti et al., 2015), the involvement of GM in the stress response psychological conditions (Kali, 2016; Mazzoli & Pessione, 2016;
is increasingly evident. This is further demonstrated by the ability Sampson & Mazmanian, 2015). The addition of GM modulation to an
to transfer stress‐prone phenotypes from one mouse to another via individual's treatment plan may be the missing link in explaining and
fecal transplantation (Collins, Kassam, & Bercik, 2013). Stress also counteracting the alarming increase in the disease burden of com‐
activates an inflammatory response via the promotion of inflamma‐ mon mental disorders.
tory cytokines (Liu, Wang, & Jiang, 2017). Given that inflammation
is recognized as underlying many psychological and neurodegen‐
2.3.1 | Lifestyle factors
erative disorders (Almond, 2013; Miller & Raison, 2016; Rea et al.,
2016), stress then appears to play a role in both the etiology and Healthy eating and exercise have long been promoted as being
maintenance of psychological illness via biological pathways, all of protective factors against both physiological and psychological
which are regulated by the GM. While this is reflective of a bot‐ conditions. Evidence suggests that one of the physiological mech‐
tom‐up process whereby GM influence stress, substantial evidence anisms through which healthy eating and exercise affect health is
also suggests the occurrence of a top‐down process through which the influence these factors have on the composition and metabolic
stress regulates GM composition (Bailey et al., 2011; Gur, Worly, & activity of GM (Mika et al., 2015; Welly et al., 2016). Essentially, a
Bailey, 2015; Knowles et al., 2008). The fact that both top‐down high‐quality diet and exercise provide the GM with the resources
and bottom‐up processes have been well established demonstrates they require to maintain optimal host function. While the influ‐
the complex cyclical and multidirectional relationship between GM, ence of diet on GM composition is widely researched, that of ex‐
stress, and psychopathology. ercise on GM receives less attention. However, exercise has been
shown to enrich microbial diversity, improve the Bacteroidetes‐to‐
Firmicutes ratio, and support the growth of SCFA‐producing bac‐
2.2.2 | Socioeconomic status
teria which have immunomodulatory effects (Monda et al., 2017).
There is an established association between low SES and several fac‐ This suggests that like diet, the beneficial outcomes of exercise
tors negatively affecting health, one of which is poor diet (Darmon may be mediated by GM which have downstream effects on men‐
& Drewnowski, 2008; Shahar, Shai, Vardi, Shahar, & Fraser, 2005). tal health.
Given that diet is the strongest environmental contributor to a prop‐
erly functioning GM (e.g., De Filippo et al., 2017; Garcia‐Mantrana
2.3.2 | Pre‐ and probiotics
et al., 2018), it is possible that the composition and/or metabolic
activities of the GM is one of the mediating factors of the relation‐ Both pre‐ and probiotics have also been demonstrated to have psy‐
ship between low SES and mental health outcomes. Another fac‐ chotropic like effects in healthy volunteers as well as those suffering
tor associated with low SES is lower educational achievement (Sirin, from conditions such as depression and chronic fatigue syndrome
2005) which may also be mediated by diet‐related changes in GM. (CFS; Akkasheh et al., 2016; Messaoudi et al., 2011; Rao et al., 2009).
Individuals who eat a poor‐quality diet (Western diet) have poorer Probiotics showing positive effects on mental health are referred
performance on cognitive tasks (Khan et al., 2015) and poorer mental to as psychobiotics (Dinan, Stanton, & Cryan, 2013). Studies have
health (Jacka, Kremer, et al., 2011; Jacka, Mykletun, Berk, Bjelland, demonstrated that various probiotic formulations (mostly including
& Tell, 2011; Markus et al., 1998) compared to those who eat high‐ Lactobacillus and Bifidobacterium species) have the ability to improve
quality diets. In addition, an association has been found between the mood in healthy (no reported diagnoses of allergic, neurological,
consumption of a Western diet and decreased left hippocampal vol‐ or psychological conditions) men and women (Benton, Williams, &
ume (Jacka, Cherbuin, Anstey, Sachdev, & Butterworth, 2015) with Brown, 2007; Messaoudi et al., 2011; Steenbergen, Sellaro, Hemert,
hippocampal volume being related to cognition (Choi et al., 2016) Bosch, & Colzato, 2015). In a placebo‐controlled study, Yamamura
and mood (Frodl et al., 2006). It is likely the GM mediate this rela‐ et al. (2009) found a probiotic formulation to improve sleep efficacy
tionship through their production of SCFAs and BDNF which have and number of awakenings (as measured by actigraphy) in an elderly
been found to be involved in neurogenesis and neuronal protection (60‐ to 81‐year‐old) sample. Moreover, a study using fMRI revealed
in mouse models (Canani, Di Costanzo, & Leone, 2012; Lee, Duan, & altered activity in brain regions responsible for emotion and sensa‐
Mattson, 2002; Sun et al., 2015). Ultimately, poorer performance on tion processing in women following four weeks of probiotic formula‐
cognitive tasks and poorer mental health limit a person's educational tion intake compared to women who received a placebo (Tillisch et
attainment (Eisenberg, Golberstein, & Hunt Justin, 2009; Fletcher, al., 2013). Also in a placebo‐controlled study, participants who took
2010; McLeod & Fettes, 2007). a prebiotic (Bimuno‐galactooligosaccharides) daily for three weeks
showed significantly lower salivary cortisol levels and decreased at‐
tentional vigilance to negative versus positive information (Schmidt
2.3 | Protective
et al., 2015). These findings were similar to those of a study that in‐
Considering the protective abilities of GM or indeed, specific mi‐ volved the administration of an SSRI (Murphy, Yiend, Lester, Cowen,
croorganisms, has the potential to revolutionize the treatment of & Harmer, 2009).
10 of 19 | GANCI et al.

of several antibiotics by increasing their inhibitory zone. As such,


2.3.3 | Fecal microbial transplant
chronic use of these drugs can induce potentially deleterious alter‐
The increasing popularity of fecal microbial transplant (FMT) in ations in GM (Macedo et al., 2017). This may partially explain treat‐
treating various conditions including but not limited to GI disorders ment resistance, although further research is needed to support this
(Brandt & Aroniadis, 2013), Parkinson's disease (Ananthaswamy, notion. Likewise, psychological interventions (e.g., cognitive behav‐
2011), autism (Aroniadis & Brandt, 2013), and ME/CFS (Borody, ior therapy) also target the brain via a focus on cognitions to affect
Nowak, & Finlayson, 2012) is perhaps due to the proliferation of behavioral change. This top‐down process has demonstrated effi‐
research associating microbial dysbiosis to a range of disorders. In cacy in the treatment of functional gastrointestinal disorders such
humans, the efficacy of FMT has been shown for conditions such as IBS (Boersma et al., 2016; Palsson & Whitehead, 2013); however,
as ulcerative colitis (Shi et al., 2016), but has not yet been demon‐ research is needed to determine whether purely psychological in‐
strated in treating psychological conditions. It does however offer terventions can enact changes in GM. By continuing to treat vari‐
a promising avenue given strong evidence suggesting a role of GM ous disorders and symptoms through pharmacological intervention
in the pathogenesis of psychological conditions (Evrensel & Ceylan, without considering the etiology of initial chemical imbalances, it is
2016). Animal models suggest that FMT is an effective way to ame‐ unlikely that rates of morbidity will decline. The net effect of ignor‐
liorate abnormal physiology and function (e.g., Sudo et al., 2004); ing etiology at the expense of treatment is therefore an increased
however, clinical trials are required to demonstrate whether this ap‐ burden upon individuals who are affected by disease, and wider
proach is equally effective in humans. Additionally, further research society.
is required into the possible risks associated with FMT. While FMT
has promising therapeutic potential, Alang and Kelly (2015) present
a case study of a patient who developed obesity following FMT 4 | R E D E FI N I N G W H O W E A R E
treatment from an overweight, but otherwise healthy donor. As GM
are associated with numerous physiological and psychological condi‐ There is currently a shift away from thinking of host–microbe interac‐
tions, FMT could theoretically result in the transference of any such tions in such binary terms toward appreciating the complexity of the
condition from donor to recipient (Bunnik, Aarts, & Chen, 2017). As relationship between the two. Binary distinctions between host and
such, it is clear that great care must be taken when screening and microbiota remain useful only in so far as to aid our understanding of
selecting potential donors. There is still much to learn about the as‐ the role of GM in psychological well‐being, which is still in its infancy.
sociations between GM and both physiological and psychological However, emerging nomenclature such as “holobiont” acknowledges
conditions, and therefore, potential long‐term risks of FMT may yet that GM are not a separate entity but are instead an integral and in‐
to emerge. separable part of human biology (Schnorr, Sankaranarayanan, Lewis,
& Warinner, 2016; Theis et al., 2016). This concept is supported by
the coevolution of humans and their microbes. The concept that the
3 | C R ITI C I S M S O F CO N V E NTI O N A L ratio of bacterial cells to human cells is approximately 1:1 (recently
TR E ATM E NT W ITH R E S PEC T TO G M revised down from previous estimations of 10:1; Sender, Fuchs, &
Milo, 2016) pays homage to the importance of respecting GM in the
Conventional treatment of psychological disorders typically in‐ conceptualization of human health and well‐being. This reconcep‐
volves pharmacological intervention such as psychotropics and/or tualization of what makes us human also provides a biological and
other medications to alter brain chemistry (e.g., Bystritsky, Khalsa, tangible basis for explaining and treating psychological illnesses that
Cameron, & Schiffman, 2013; Lieberman et al., 2005). Although ben‐ can often be considered abstract.
eficial, such treatments may induce undesirable side effects includ‐
ing, but not limited to, nausea, sleep disturbance, weight gain, and
sexual dysfunction (Ferguson, 2001) all of which may themselves 5 | C H A LLE N G E S A N D TH E WAY
be a result of microbe‐mediated drug metabolism (Enright, Joyce, FO RWA R D
Gahan, & Griffin, 2017). Despite the ever increasing reliance on phar‐
macotherapy (Kallivayalil, 2008; Vozeh, 2003), disease states remain Understanding the multidirectional relationship between GM and
relatively stable which suggests the need for auxiliary treatment op‐ the nervous system is hindered by its inherent complexity (Mazzoli
tions and/or targets which take into account several body systems, & Pessione, 2016). However, while still in its infancy, interdiscipli‐
including the GM. Many psychotropic drugs, known for their influ‐ nary research has uncovered novel ways of conceptualizing disease.
ence on CNS receptor function, also demonstrate antimicrobial ef‐ While theoretically relevant, research into the BGMA also has im‐
fects (Kalayci, Demirci, & Sahin, 2014) which may have unintended portant practical implications, offering a more holistic approach to
consequences on the BGMA. This is particularly true of many SSRIs treatment and prevention of psychological illness. While this new
commonly used to treat depression and anxiety disorders. field of research is promising, it remains unclear which factors, and
In addition to having direct antimicrobial effects, Ayaz et al. in which combination, alter the balance between symptomatic and
(2015) found that sertraline (an SSRI) augments the effectiveness asymptomatic outcomes.
GANCI et al. | 11 of 19

The sheer number of confounding variables makes it difficult and potentially even FMT in the treatment plans of their clients, in
to establish causational links between GM and symptomatology. conjunction with conventional psychological treatments.
However, as understanding of the GM advances, so too will research It is not the contention of this paper to claim that ameliorating
methodology and technology. It is only with continued research into gut health is the panacea to all psychological disorders and symp‐
the link between GM and psychological illness that we will be able to tomatology. Instead, it is to demonstrate the complex intercon‐
elucidate the true extent to which our resident microbes contribute nectivity between multiple body systems in disease processes,
to mental health. As the majority of studies regarding the role of from etiology through to treatment, and ideally prevention. In
GM in both physiological and psychological health and disease have support of the call to action by Allen et al. (2017), the discipline of
been conducted using animal models, clinical trials with human sam‐ psychology must shift away from its CNS‐centric conceptualiza‐
ples are imperative to the advancement of knowledge and ultimately tion of disease and symptom‐centered disease treatment models
practical application. toward a multidisciplinary approach to treatment and prevention.
As the burgeoning research into the relationship between GM This paradigm shift will empower psychologists to better treat and
and psychological health outcomes gathers momentum, so too care for their clients. A multidisciplinary approach where health‐
does the call to action for psychologists to embrace the microbi‐ care professionals across a variety of disciplines have a united ap‐
ome as a potential factor in explaining, treating, and preventing proach to treatment and prevention will also empower the public
mental illness. This is an important paradigm shift which must to better understand and take control of their physiological and
occur within the discipline of psychology in order to keep up to psychological health. It is only through this shared awareness that
date with the most current and complete knowledge of the human the healthcare community can make inroads into improving the
body and mind which translates into providing the best possible mental health of current and future generations.
care for clients. While it is unnecessary and impractical to expect
psychologists to develop a detailed understanding of the influence C O N FL I C T O F I N T E R E S T
of GM on mental health, it is important that the role of the GM is
None declared.
acknowledged, especially in the absence of clear social and emo‐
tional factors. Facilitating this paradigm shift may require change
at a “grass roots” level, where psychobiology is better integrated DATA AVA I L A B I L I T Y S TAT E M E N T
into higher education psychology degrees. Additionally, profes‐
Data sharing is not applicable to this article as no new data were cre‐
sional development courses regarding the role of the GM in mental
ated or analyzed in this study.
health should be established and promoted to current practicing
psychologists who can use this information to provide more com‐
ORCID
plete care for their clients.
The fact that GM are able to influence psychological functioning Michael Ganci https://orcid.org/0000-0003-2815-0548
is an exciting and encouraging prospect, which begs for multidisci‐
plinary approaches to both research and practice. In terms of prac‐
REFERENCES
tical implications, increasing our understanding of the mechanisms
that mediate communication processes between GM and host has Abdou, A. M., Higashiguchi, S., Horie, K., Kim, M., Hatta, H., & Yokogoshi,
the potential to inform strategies to limit the damaging aspects of H. (2006). Relaxation and immunity enhancement effects of gamma‐
aminobutyric acid (GABA) administration in humans. BioFactors,
this communication. This will provide new avenues of treatment for a
26(3), 201–208.
wide range of symptoms and disorders (Freestone, 2013) as well as to
Abeles, S. R., Jones, M. B., Santiago‐Rodriguez, T. M., Ly, M., Klitgord,
promote good health. This will require a substantial shift away from N., Yooseph, S., … Pride, D. T. (2016). Microbial diversity in in‐
the reductionist approaches that see us working exclusively in our dividuals and their household contacts following typical anti‐
specific field. This is not to suggest that psychologists should become biotic courses. Microbiome, 4(1), 39. https​ ://doi.org/10.1186/
s40168-016-0187-9
expert in areas outside of their field, but instead to understand and ac‐
Akkasheh, G., Kashani‐Poor, Z., Tajabadi‐Ebrahimi, M., Jafari, P., Akbari,
knowledge that the best way forward is a multidisciplinary approach H., Taghizadeh, M., … Esmaillzadeh, A. (2016). Clinical and meta‐
to the treatment and prevention of mental illness. A shift toward a bolic response to probiotic administration in patients with major
multidisciplinary, and therefore more holistic, approach will provide depressive disorder: A randomized, double‐blind, placebo‐con‐
trolled trial. Nutrition, 32(3), 315–320. https​ ://doi.org/10.1016/j.
an opportunity to better understand the etiology of disease which
nut.2015.09.003
requires the expertise of several disciplines and a consideration of Alang, N., & Kelly, C. R. (2015). Weight gain after fecal microbiota trans‐
key body systems, including the GM, as intertwined and inseparable. plantation. Open Forum Infectious Diseases, 2(1), ofv004. https​://doi.
In light of the evidence research has provided thus far, psychologists org/10.1093/ofid/ofv004
Alcock, J., Maley, C. C., & Aktipis, C. A. (2014). Is eating behavior ma‐
working as part of multidisciplinary teams with other professionals
nipulated by the gastrointestinal microbiota? Evolutionary pressures
such as nutritionists, gastroenterologists, and microbiologists must and potential mechanisms. BioEssays, 36(10), 940–949. https​://doi.
seriously consider the inclusion of dietary plans, pre‐ and probiotics, org/10.1002/bies.20140 ​0 071
12 of 19 | GANCI et al.

Alkasir, R., Li, J., Li, X., Jin, M., & Zhu, B. (2017). Human gut microbiota: Benton, D., Williams, C., & Brown, A. (2007). Impact of consuming a
The links with dementia development. Protein Cell, 8(2), 90–102. milk drink containing a probiotic on mood and cognition. European
https​://doi.org/10.1007/s13238-016-0338-6 Journal of Clinical Nutrition, 61(3), 355–361. https​://doi.org/10.1038/
Allen, A. P., Dinan, T. G., Clarke, G., & Cryan, J. F. (2017). A psychol‐ sj.ejcn.1602546
ogy of the human brain‐gut‐microbiome axis. Social and Personality Bercik, P., Park, A. J., Sinclair, D., Khoshdel, A., Lu, J., Huang, X., …
Psychology Compass, 11(4), e12309. https​ ://doi.org/10.1111/ Verdu, E. F. (2011). The anxiolytic effect of Bifidobacterium longum
spc3.12309​ NCC3001 involves vagal pathways for gut‐brain communication.
Allen, B., & Nowak, M. A. (2013). Cooperation and the fate of microbial Neurogastroenterology and Motility, 23(12), 1132–1139. https​ ://doi.
societies. PLoS Biology, 11(4), e1001549. https​ ://doi.org/10.1371/ org/10.1111/j.1365-2982.2011.01796.x
journ​al.pbio.1001549 Berger, M., Juster, R. P., & Sarnyai, Z. (2015). Mental health consequences
Almond, M. (2013). Depression and inflammation: Examining the link. of stress and trauma: Allostatic load markers for practice and policy
Current Psychiatry, 12(6), 24–32. with a focus on Indigenous health. Australas Psychiatry, 23(6), 644–
Ananthaswamy, A. (2011). Faecal transplant eases symptoms of 649. https​://doi.org/10.1177/10398​56215​608281
Parkinson's disease. New Journal of Science, 209, 8–9. Berkes, J., Viswanathan, V. K., Savkovic, S. D., & Hecht, G. (2003).
Anderson, G., & Maes, M. (2015). The gut–brain axis: The role of mel‐ Intestinal epithelial responses to enteric pathogens: Effects on the
atonin in linking psychiatric, inflammatory and neurodegenerative tight junction barrier, ion transport, and inflammation. Gut, 52(3),
conditions. Advances in Integrative Medicine, 2(1), 31–37. https​://doi. 439–451. https​://doi.org/10.1136/gut.52.3.439
org/10.1016/j.aimed.2014.12.007 Biagi, E., Nylund, L., Candela, M., Ostan, R., Bucci, L., Pini, E., … De Vos,
Anderson, R. C., Cookson, A. L., McNabb, W. C., Park, Z., McCann, M. W. (2010). Through ageing, and beyond: Gut microbiota and inflam‐
J., Kelly, W. J., & Roy, N. C. (2010). Lactobacillus plantarum MB452 matory status in seniors and centenarians. PLoS ONE, 5(5), e10667.
enhances the function of the intestinal barrier by increasing the ex‐ https​://doi.org/10.1371/journ​al.pone.0010667
pression levels of genes involved in tight junction formation. BMC Bischoff, S. C., Barbara, G., Buurman, W., Ockhuizen, T., Schulzke, J.‐D.,
Microbiology, 10, 316. https​://doi.org/10.1186/1471-2180-10-316 Serino, M., … Wells, J. M. (2014). Intestinal permeability – a new tar‐
Andreas, S., Schulz, H., Volkert, J., Dehoust, M., Sehner, S., Suling, A., … get for disease prevention and therapy. BMC Gastroenterology, 14(1),
Härter, M. (2017). Prevalence of mental disorders in elderly people: 189. https​://doi.org/10.1186/s12876-014-0189-7
The European MentDis_ICF65+ study. The British Journal of Psychiatry, Blumstein, D. T., Levy, K., Mayer, E., & Harte, J. (2014). Gastrointestinal
210(2), 125–131. https​://doi.org/10.1192/bjp.bp.115.180463 dysbiosis. Evolution, Medicine, and Public Health, 2014(1), 163. https​://
Angst, J., Merikangas, K. R., & Preisig, M. (1997). Subthreshold syn‐ doi.org/10.1093/emph/eou029
dromes of depression and anxiety in the community. Journal of Boersma, K., Ljótsson, B., Edebol‐Carlman, H., Schrooten, M., Linton,
Clinical Psychiatry, 58(Suppl 8), 6–10. S. J., & Brummer, R. J. (2016). Exposure‐based cognitive behavioral
Anisman, H., & Zacharko, R. (1982). Depression: The predisposing influ‐ therapy for irritable bowel syndrome. A single‐case experimental de‐
ence of stress. Behavioral and Brain Sciences, 5(1), 89–99. https​://doi. sign across 13 subjects. Cognitive Behaviour Therapy, 45(6), 415–430.
org/10.1017/S0140​525X0​0 010633 https​://doi.org/10.1080/16506​073.2016.1194455
Arentsen, T., Raith, H., Qian, Y., Forssberg, H., & Diaz Heijtz, R. (2015). Boonstra, E., de Kleijn, R., Colzato, L. S., Alkemade, A., Forstmann, B. U.,
Host microbiota modulates development of social preference in & Nieuwenhuis, S. (2015). Neurotransmitters as food supplements:
mice. Microbial Ecology in Health & Disease, 26, 29719. https​://doi. The effects of GABA on brain and behavior. Frontiers in Psychology, 6,
org/10.3402/mehd.v26.29719​ 1520. https​://doi.org/10.3389/fpsyg.2015.01520​
Aroniadis, O. C., & Brandt, L. J. (2013). Fecal microbiota transplantation: Borody, T. J., Nowak, A., & Finlayson, S. (2012). The GI microbiome and
Past, present and future. Current Opinion in Gastroenterology, 29(1), its role in chronic fatigue syndrome: A summary of bacteriotherapy.
79–84. https​://doi.org/10.1097/MOG.0b013​e3283​5a4b3e Journal of the Australasian College of Nutritional and Environmental
Arreola, R., Becerril‐Villanueva, E., Cruz‐Fuentes, C., Velasco‐ Medicine, 31(3), 3–8.
Velázquez, M. A., Garcés‐Alvarez, M. E., Hurtado‐Alvarado, G., … Borre, Y. E., O'Keeffe, G. W., Clarke, G., Stanton, C., Dinan, T. G., &
Pavón, L. (2015). Immunomodulatory effects mediated by sero‐ Cryan, J. F. (2014). Microbiota and neurodevelopmental windows:
tonin. Journal of Immunology Research, 2015, 354957. https​ ://doi. Implications for brain disorders. Trends in Molecular Medicine, 20(9),
org/10.1155/2015/354957 509–518. https​://doi.org/10.1016/j.molmed.2014.05.002
Asnicar, F., Manara, S., Zolfo, M., Truong, D. T., Scholz, M., Armanini, F., Bourassa, M. W., Alim, I., Bultman, S. J., & Ratan, R. R. (2016). Butyrate,
… Segata, N. (2017). Studying vertical microbiome transmission from neuroepigenetics and the gut microbiome: Can a high fiber diet im‐
mothers to infants by strain‐level metagenomic profiling. mSystems, prove brain health? Neuroscience Letters, 625, 56–63. https​://doi.
2(1), e00164–16. https​://doi.org/10.1128/mSyst​ems.00164-16 org/10.1016/j.neulet.2016.02.009
Ayaz, M., Subhan, F., Ahmed, J., Khan, A.‐U., Ullah, F., Ullah, I., … Hussain, Brandt, L. J., & Aroniadis, O. C. (2013). An overview of fecal micro‐
S. (2015). Sertraline enhances the activity of antimicrobial agents biota transplantation: Techniques, indications, and outcomes.
against pathogens of clinical relevance. Journal of Biological Research‐ Gastrointestinal Endoscopy, 78(2), 240–249. https​://doi.org/10.1016/j.
Thessaloniki, 22(1), 4. https​://doi.org/10.1186/s40709-015-0028-1 gie.2013.03.1329
Bailey, M. T., Dowd, S. E., Galley, J. D., Hufnagle, A. R., Allen, R. G., & Braniste, V., Al‐Asmakh, M., Kowal, C., Anuar, F., Abbaspour, A., Toth, M.,
Lyte, M. (2011). Exposure to a social stressor alters the structure of … Pettersson, S. (2014). The gut microbiota influences blood‐brain
the intestinal microbiota: Implications for stressor‐induced immuno‐ barrier permeability in mice. Science Translational Medicine, 6(263),
modulation. Brain, Behavior, and Immunity, 25(3), 397–407. https​:// 263ra158. https​://doi.org/10.1126/scitr​anslm​ed.3009759
doi.org/10.1016/j.bbi.2010.10.023 Bravo, J. A., Forsythe, P., Chew, M. V., Escaravage, E., Savignac, H. M.,
Ballou, S., Bedell, A., & Keefer, L. (2015). Psychosocial impact of irrita‐ Dinan, T. G., … Cryan, J. F. (2011). Ingestion of Lactobacillus strain
ble bowel syndrome: A brief review. World Journal of Gastrointestinal regulates emotional behavior and central GABA receptor expression
Pathophysiology, 6(4), 120–123. https​ ://doi.org/10.4291/wjgp. in a mouse via the vagus nerve. Proceedings of the National Academy
v6.i4.120 of Sciences USA, 108(38), 16050–16055. https​ ://doi.org/10.1073/
Behary, P., & Miras, A. D. (2015). Food preferences and underlying mech‐ pnas.11029​99108​
anisms after bariatric surgery. The Proceedings of the Nutrition Society, Broussard, J. L., & Devkota, S. (2016). The changing microbial land‐
74(4), 419–425. https​://doi.org/10.1017/s0029​66511​5002074 scape of Western society: Diet, dwellings and discordance.
GANCI et al. | 13 of 19

Molecular Metabolism, 5(9), 737–742. https​ ://doi.org/10.1016/j. Collins, S. M., Kassam, Z., & Bercik, P. (2013). The adoptive transfer of
molmet.2016.07.007 behavioral phenotype via the intestinal microbiota: Experimental ev‐
Buford, T. W. (2017). (Dis)Trust your gut: The gut microbiome in age‐re‐ idence and clinical implications. Current Opinion in Microbiology, 16(3),
lated inflammation, health, and disease. Microbiome, 5(1), 80. https​:// 240–245. https​://doi.org/10.1016/j.mib.2013.06.004
doi.org/10.1186/s40168-017-0296-0 Corcoran, C., Walker, E., Huot, R., Mittal, V., Tessner, K., Kestler, L., &
Bunnik, E. M., Aarts, N., & Chen, L. A. (2017). Physicians must discuss po‐ Malaspina, D. (2003). The stress cascade and schizophrenia: Etiology
tential long‐term risks of fecal microbiota transplantation to ensure and onset. Schizophrenia Bulletin, 29(4), 671–692. https​ ://doi.
informed consent. The American Journal of Bioethics, 17(5), 61–63. org/10.1093/oxfor​djour​nals.schbul.a007038
https​://doi.org/10.1080/15265​161.2017.1299816 Cox, L. M., Yamanishi, S., Sohn, J., Alekseyenko, A. V., Leung, J. M., Cho,
Burch, J. D. (2016). Intestinal infection associated with future onset of an I., … Blaser, M. J. (2014). Altering the intestinal microbiota during a
anxiety disorder: Results of a nationally representative study. Brain, critical developmental window has lasting metabolic consequences.
Behaviour, and Immunity, 57, 222–226. https​ ://doi.org/10.1016/j. Cell, 158(4), 705–721. https​://doi.org/10.1016/j.cell.2014.05.052
bbi.2016.05.014 Craig, A. D. (2002). How do you feel? Interoception: The sense of the
Byrne, C. S., Chambers, E. S., Morrison, D. J., & Frost, G. (2015). The role physiological condition of the body. Nature Reviews Neuroscience,
of short chain fatty acids in appetite regulation and energy homeo‐ 3(8), 655–666. https​://doi.org/10.1038/nrn894
stasis. International Journal of Obesity, 39(9), 1331–1338. https​://doi. Cryan, J. F., & Dinan, T. G. (2012). Mind‐altering microorganisms: The
org/10.1038/ijo.2015.84 impact of the gut microbiota on brain and behaviour. Nature Reviews
Bystritsky, A., Khalsa, S. S., Cameron, M. E., & Schiffman, J. (2013). Neuroscience, 13(10), 701–712. https​://doi.org/10.1038/nrn3346
Current diagnosis and treatment of anxiety disorders. Pharmacology Cryan, J. F., & O'Mahony, S. M. (2011). The microbiome‐gut‐brain axis:
& Therapeutics, 38(1), 30–57. From bowel to behavior. Neurogastroenterology and Motility, 23(3),
Campell, A. W. (2014). Autoimmunity and the gut. Autoimmune Diseases, 187–192. https​://doi.org/10.1111/j.1365-2982.2010.01664.x
2014, 1–12. https​://doi.org/10.1155/2014/152428 Cui, L., Zhao, T., Hu, H., Zhang, W., & Hua, X. (2017). Association study
Canani, R. B., Di Costanzo, M., & Leone, L. (2012). The epigenetic effects of gut flora in coronary heart disease through high‐throughput se‐
of butyrate: Potential therapeutic implications for clinical practice. quencing. BioMed Research International, 2017, 3796359. https​://doi.
Clinical Epigenetics, 4(1), 4. https​://doi.org/10.1186/1868-7083-4-4 org/10.1155/2017/3796359
Canavan, C., West, J., & Card, T. (2015). Change in quality of life for pa‐ Dąbrowska, K., & Witkiewicz, W. (2016). Correlations of host genetics
tients with irritable bowel syndrome following referral to a gastroen‐ and gut microbiome composition. Frontiers in Microbiology, 7, 1357.
terologist: A cohort study. PLoS ONE, 10(10), e0139389. https​://doi. https​://doi.org/10.3389/fmicb.2016.01357​
org/10.1371/journ​al.pone.0139389 Darmon, N., & Drewnowski, A. (2008). Does social class predict diet
Cani, P. D., Lecourt, E., Dewulf, E. M., Sohet, F. M., Pachikian, B. D., quality? American Journal of Clinical Nutrition, 87(5), 1107–1117. https​
Naslain, D., … Delzenne, N. M. (2009). Gut microbiota fermentation ://doi.org/10.1093/ajcn/87.5.1107
of prebiotics increases satietogenic and incretin gut peptide produc‐ David, L. A., Maurice, C. F., Carmody, R. N., Gootenberg, D. B., Button, J.
tion with consequences for appetite sensation and glucose response E., Wolfe, B. E., … Turnbaugh, P. J. (2014). Diet rapidly and reproduc‐
after a meal. American Journal of Clinical Nutrition, 90(5), 1236–1243. ibly alters the human gut microbiome. Nature, 505(7484), 559–563.
https​://doi.org/10.3945/ajcn.2009.28095​ https​://doi.org/10.1038/natur​e12820
Carabotti, M., Scirocco, A., Maselli, M. A., & Severi, C. (2015). The gut‐ De Filippo, C., Di Paola, M., Ramazzotti, M., Albanese, D., Pieraccini, G.,
brain axis: Interactions between enteric microbiota, central and en‐ Banci, E., … Lionetti, P. (2017). Diet, environments, and gut microbi‐
teric nervous systems. Annals of Gastroenterology, 28(2), 203–209. ota: A preliminary investigation in children living in rural and urban
Castro‐Nallar, E., Bendall, M. L., Pérez‐Losada, M., Sabuncyan, Burkina Faso and Italy. Frontiers in Microbiology, 8, 1979. https​://doi.
S., Severance, E. G., Dickerson, F. B., … Crandall, K. A. (2015). org/10.3389/fmicb.2017.01979​
Composition, taxonomy and functional diversity of the oropharynx DeGruttola, A. K., Low, D., Mizoguchi, A., & Mizoguchi, E. (2016).
microbiome in individuals with schizophrenia and controls. PeerJ, 3, Current understanding of dysbiosis in disease in human and animal
e1140. https​://doi.org/10.7717/peerj.1140 models. Inflammatory Bowel Diseases, 22(5), 1137–1150. https​://doi.
Chambers, E. S., Viardot, A., Psichas, A., Morrison, D. J., Murphy, K. G., org/10.1097/MIB.00000​0 0000​0 00750
Zac‐Varghese, S. E. K., … Frost, G. (2015). Effects of targeted deliv‐ den Besten, G., van Eunen, K., Groen, A. K., Venema, K., Reijngoud, D.
ery of propionate to the human colon on appetite regulation, body J., & Bakker, B. M. (2013). The role of short‐chain fatty acids in the
weight maintenance and adiposity in overweight adults. Gut, 64(11), interplay between diet, gut microbiota, and host energy metabolism.
1744–1754. https​://doi.org/10.1136/gutjnl-2014-307913 Journal of Lipid Research, 54(9), 2325–2340. https​://doi.org/10.1194/
Cherlyn, S. Y., Woon, P. S., Liu, J. J., Ong, W. Y., Tsai, G. C., & Sim, K. jlr.R036012
(2010). Genetic association studies of glutamate, GABA and related Desbonnet, L., Garrett, L., Clarke, G., Kiely, B., Cryan, J. F., & Dinan, T. G.
genes in schizophrenia and bipolar disorder: A decade of advance. (2010). Effects of the probiotic Bifidobacterium infantis in the ma‐
Neuroscience and Biobehavioral Reviews, 34(6), 958–977. https​://doi. ternal separation model of depression. Neuroscience, 170(4), 1179–
org/10.1016/j.neubi​orev.2010.01.002 1188. https​://doi.org/10.1016/j.neuro​scien​ce.2010.08.005
Choi, M.‐H., Kim, H.‐S., Gim, S.‐Y., Kim, W.‐R., Mun, K.‐R., Tack, G.‐R., … Di Chiara, G., & Bassareo, V. (2007). Reward system and addiction: What
Chung, S.‐C. (2016). Differences in cognitive ability and hippocampal dopamine does and doesn't do. Current Opinion in Pharmacology, 7(1),
volume between Alzheimer's disease, amnestic mild cognitive impair‐ 69–76. https​://doi.org/10.1016/j.coph.2006.11.003
ment, and healthy control groups, and their correlation. Neuroscience Dinan, T. G., Stanton, C., & Cryan, J. F. (2013). Psychobiotics: A novel
Letters, 620, 115–120. https​://doi.org/10.1016/j.neulet.2016.03.044 class of psychotropic. Biological Psychiatry, 74(10), 720–726. https​://
Christensen, L., & Brooks, A. (2006). Changing food preference as a doi.org/10.1016/j.biops​ych.2013.05.001
function of mood. Journal of Psychology, 140(4), 293–306. https​:// Eisenberg, D., Golberstein, E., & Hunt Justin, B. (2009). Mental health
doi.org/10.3200/jrlp.140.4.293-306 and academic success in college. The B.E. Journal of Economic Analysis
Clarke, G., Stilling, R. M., Kennedy, P. J., Stanton, C., Cryan, J. F., & Dinan, & Policy, 9(1), 1–37. https​://doi.org/10.2202/1935-1682.2191
T. G. (2014). Minireview: Gut microbiota: The neglected endocrine Eisenhofer, G., Åneman, A., Friberg, P., Hooper, D., Fåndriks, L., Lonroth,
organ. Molecular Endocrinology, 28(8), 1221–1238. https​ ://doi. H., … Mezey, E. (1997). Substantial production of dopamine in the
org/10.1210/me.2014-1108 human gastrointestinal tract. The Journal of Clinical Endocrinology
14 of 19 | GANCI et al.

& Metabolism, 82(11), 3864–3871. https​ ://doi.org/10.1210/ Frodl, T., Schaub, A., Banac, S., Charypar, M., Jäger, M., Kümmler, P., …
jcem.82.11.4339 Meisenzahl, E. M. (2006). Reduced hippocampal volume correlates
Emoto, T., Yamashita, T., Kobayashi, T., Sasaki, N., Hirota, Y., Hayashi, T., with executive dysfunctioning in major depression. Journal of
… Hirata, K.‐I. (2017). Characterization of gut microbiota profiles in Psychiatry and Neuroscience, 31(5), 316–325.
coronary artery disease patients using data mining analysis of termi‐ Furet, J.‐P., Kong, L.‐C., Tap, J., Poitou, C., Basdevant, A., Bouillot, J.‐L., …
nal restriction fragment length polymorphism: Gut microbiota could Clement, K. (2010). Differential adaptation of human gut microbiota
be a diagnostic marker of coronary artery disease. Heart and Vessels, to bariatric surgery‐induced weight loss: Links with metabolic and
32(1), 39–46. https​://doi.org/10.1007/s00380-016-0841-y low‐grade inflammation markers. Diabetes, 59(12), 3049–3057. https​
Enright, E. F., Joyce, S. A., Gahan, C. G., & Griffin, B. T. (2017). Impact ://doi.org/10.2337/db10-0253
of gut microbiota‐mediated bile acid metabolism on the solubi‐ Furusawa, Y., Obata, Y., Fukuda, S., Endo, T. A., Nakato, G., Takahashi,
lization capacity of bile salt micelles and drug solubility. Molecular D., … Ohno, H. (2013). Commensal microbe‐derived butyrate induces
Pharmaceutics, 14(4), 1251–1263. https​://doi.org/10.1021/acs.molph​ the differentiation of colonic regulatory T cells. Nature, 504(7480),
armac​eut.6b01155 446–450. https​://doi.org/10.1038/natur​e12721
Evrensel, A., & Ceylan, M. E. (2015). The gut‐brain axis: The missing link Gagniere, J., Raisch, J., Veziant, J., Barnich, N., Bonnet, R., Buc, E., …
in depression. Clinical Psychopharmacology and Neuroscience, 13(3), Bonnet, M. (2016). Gut microbiota imbalance and colorectal can‐
239–244. https​://doi.org/10.9758/cpn.2015.13.3.239 cer. World Journal of Gastroenterology, 22(2), 501–518. https​://doi.
Evrensel, A., & Ceylan, M. E. (2016). Fecal microbiota transplantation and org/10.3748/wjg.v22.i2.501
its usage in neuropsychiatric disorders. Clinical Psychopharmacology Garakani, A., Win, T., Virk, S., Gupta, S., Kaplan, D., & Masand, P. S.
and Neuroscience, 14(3), 231–237. https​ ://doi.org/10.9758/ (2003). Comorbidity of irritable bowel syndrome in psychiatric pa‐
cpn.2016.14.3.231 tients: A review. American Journal of Therapeutics, 10(1), 61–67. https​
Fasano, A. (2012). Intestinal permeability and its regulation by zonulin: ://doi.org/10.1097/00045​391-20030​1000-00014​
Diagnostic and therapeutic implications. Clinical Gastroenterology Garcia‐Mantrana, I., Selma‐Royo, M., Alcantara, C., & Collado, M. C.
and Hepatology, 10(10), 1096–1100. https​ ://doi.org/10.1016/j. (2018). Shifts on gut microbiota associated to mediterranean diet
cgh.2012.08.012 adherence and specific dietary intakes on general adult popula‐
Femenía, T., Gómez‐Galán, M., Lindskog, M., & Magara, S. (2012). tion. Frontiers in Microbiology, 9, 890. https​ ://doi.org/10.3389/
Dysfunctional hippocampal activity affects emotion and cogni‐ fmicb.2018.00890​
tion in mood disorders. Brain Research, 1476, 58–70. https​ ://doi. Garrett, W. S. (2015). Cancer and the microbiota. Science, 348(6230),
org/10.1016/j.brain​res.2012.03.053 80–86. https​://doi.org/10.1126/scien​ce.aaa4972
Ferguson, E. (2013). Personality is of central concern to under‐ Ghorbani, A., Salari, M., Shaygannejad, V., & Norouzi, R. (2013). The role
stand health: Towards a theoretical model for health psychol‐ of melatonin in the pathogenesis of multiple sclerosis: A case‐con‐
ogy. Health Psychology Review, 7(Suppl 1), S32–S70. https​ ://doi. trol study. International Journal of Preventive Medicine, 4(Suppl 2),
org/10.1080/17437​199.2010.547985 S180–S184.
Ferguson, J. M. (2001). SSRI antidepressant medications: adverse effects Giunta, B., Fernandez, F., Nikolic, W. V., Obregon, D., Rrapo, E., Town,
and tolerability. Primary Care Companion to the Journal of Clinical T., & Tan, J. (2008). Inflammaging as a prodrome to Alzheimer's
Psychiatry, 3(1), 22–27. https​://doi.org/10.4088/PCC.v03n0105 disease. Journal of Neuroinflammation, 5, 51. https​ ://doi.
Finegold, S. M., Molitoris, D., Song, Y., Liu, C., Vaisanen, M.‐L., Bolte, E., org/10.1186/1742-2094-5-51
… Kaul, A. (2002). Gastrointestinal microflora studies in late‐onset Glanz, K., Basil, M., Maibach, E., Goldberg, J., & Snyder, D. (1998). Why
autism. Clinical Infectious Diseases, 35(Suppl 1), S6–s16. https​://doi. Americans eat what they do: Taste, nutrition, cost, convenience, and
org/10.1086/341914 weight control concerns as influences on food consumption. Journal
Fiorentino, M., Sapone, A., Senger, S., Camhi, S. S., Kadzielski, S. M., of the American Dietetic Association, 98(10), 1118–1126. https​://doi.
Buie, T. M., … Fasano, A. (2016). Blood‐brain barrier and intestinal org/10.1016/s0002-8223(98)00260-0
epithelial barrier alterations in autism spectrum disorders. Molecular Gluckman, P. D., Low, F. M., Buklijas, T., Hanson, M. A., & Beedle, A. S.
Autism, 7, 49. https​://doi.org/10.1186/s13229-016-0110-z (2011). How evolutionary principles improve the understanding of
Fletcher, J. M. (2010). Adolescent depression and educational attain‐ human health and disease. Evolutionary Applications, 4(2), 249–263.
ment: Results using sibling fixed effects. Health Economics, 19(7), https​://doi.org/10.1111/j.1752-4571.2010.00164.x
855–871. https​://doi.org/10.1002/hec.1526 Goodrich, J. K., Waters, J. L., Poole, A. C., Sutter, J. L., Koren, O., Blekhman,
Fornaro, M., Prestia, D., Colicchio, S., & Perugi, G. (2010). A systematic, R., … Ley, R. E. (2014). Human genetics shape the gut microbiome.
updated review on the antidepressant agomelatine focusing on its Cell, 159(4), 789–799. https​://doi.org/10.1016/j.cell.2014.09.053
melatonergic modulation. Current Neuropharmacology, 8(3), 287– Grace, A. A. (2016). Dysregulation of the dopamine system in
304. https​://doi.org/10.2174/15701​59107​92246227 the pathophysiology of schizophrenia and depression. Nature
Franceschi, C., Capri, M., Monti, D., Giunta, S., Olivieri, F., Sevini, F., … Reviews Neuroscience, 17(8), 524–532. https​ ://doi.org/10.1038/
Salvioli, S. (2007). Inflammaging and anti‐inflammaging: A systemic nrn.2016.57
perspective on aging and longevity emerged from studies in humans. Grenham, S., Clarke, G., Cryan, J. F., & Dinan, T. G. (2011). Brain‐gut‐mi‐
Mechanisms of Ageing and Development, 128(1), 92–105. https​://doi. crobe communication in health and disease. Frontiers in Physiology, 2,
org/10.1016/j.mad.2006.11.016 94. https​://doi.org/10.3389/fphys.2011.00094​
Freestone, P. (2013). Communication between bacteria and their hosts. Gulesserian, T., Engidawork, E., Cairns, N., & Lubec, G. (2000). Increased
Scientifica, 2013, 1–15. https​://doi.org/10.1155/2013/361073 protein levels of serotonin transporter in frontal cortex of patients
Fremont, M., Coomans, D., Massart, S., & De Meirleir, K. (2013). High‐ with Down syndrome. Neuroscience Letters, 296(1), 53–57. https​://
throughput 16S rRNA gene sequencing reveals alterations of in‐ doi.org/10.1016/S0304-3940(00)01624-4
testinal microbiota in myalgic encephalomyelitis/chronic fatigue Gur, T. L., Worly, B. L., & Bailey, M. T. (2015). Stress and the commen‐
syndrome patients. Anaerobe, 22, 50–56. https​://doi.org/10.1016/j. sal microbiota: Importance in parturition and infant neurode‐
anaer​obe.2013.06.002 velopment. Frontiers in Psychiatry, 6, 5. https​ ://doi.org/10.3389/
Friedrich, M. (2017). Depression is the leading cause of disability fpsyt.2015.00005​
around the world. JAMA, 317(15), 1517. https​ ://doi.org/10.1001/ Hakansson, A., & Molin, G. (2011). Gut microbiota and inflammation.
jama.2017.3826 Nutrients, 3(6), 637–682. https​://doi.org/10.3390/nu306​0637
GANCI et al. | 15 of 19

Halford, J. C., & Blundell, J. E. (2000). Separate systems for serotonin and Jeffery, I. B., O'Toole, P. W., Ohman, L., Claesson, M. J., Deane, J., Quigley,
leptin in appetite control. Annals of Medicine, 32(3), 222–232. https​:// E. M., & Simren, M. (2012). An irritable bowel syndrome subtype de‐
doi.org/10.3109/07853​89000​8998829 fined by species‐specific alterations in faecal microbiota. Gut, 61(7),
Haller, H., Cramer, H., Lauche, R., Gass, F., & Dobos, G. J. (2014). The 997–1006. https​://doi.org/10.1136/gutjnl-2011-301501
prevalence and burden of subthreshold generalized anxiety disor‐ Jenkins, T. A., Nguyen, J. C., Polglaze, K. E., & Bertrand, P. P. (2016).
der: A systematic review. BMC Psychiatry, 14(128), 1–13. https​://doi. Influence of tryptophan and serotonin on mood and cognition with
org/10.1186/1471-244X-14-128 a possible role of the gut‐brain axis. Nutrients, 8(1), 56. https​://doi.
Harvey, S. B., Deady, M., Wang, M., Mykletun, A., Butterworth, P., org/10.3390/nu801​0 056
Christensen, H., & Mitchell, P. B. (2017). Is the prevalence of mental Jiang, H., Ling, Z., Zhang, Y., Mao, H., Ma, Z., Yin, Y., … Ruan, B. (2015).
illness increasing in Australia? Evidence from national health surveys Altered fecal microbiota composition in patients with major depres‐
and administrative data, 2001–2004. Medical Journal of Australia, sive disorder. Brain, Behavior, and Immunity, 48, 186–194. https​://doi.
206(11), 490–493. https​://doi.org/10.5694/mja16.00295​ org/10.1016/j.bbi.2015.03.016
Heiman, M. L., & Greenway, F. L. (2016). A healthy gastrointestinal micro‐ Johansen, T., & Dittrich, W. H. (2013). Cognitive performance in a subclin‐
biome is dependent on dietary diversity. Molecular Metabolism, 5(5), ical obsessive‐compulsive sample 1: Cognitive functions. Psychiatry
317–320. https​://doi.org/10.1016/j.molmet.2016.02.005 Journal, 1, 1–10. https​://doi.org/10.1155/2013/565191
Hibbing, M. E., Fuqua, C., Parsek, M. R., & Peterson, S. B. (2010). Bacterial Jumbe, S., Hamlet, C., & Meyrick, J. (2017). Psychological aspects of bar‐
competition: Surviving and thriving in the microbial jungle. Nature iatric surgery as a treatment for obesity. Current Obesity Reports, 6(1),
Reviews Microbiology, 8(1), 15–25. https​ ://doi.org/10.1038/nrmic​ 71–78. https​://doi.org/10.1007/s13679-017-0242-2
ro2259 Kalayci, S., Demirci, S., & Sahin, F. (2014). Determination of antimicro‐
Holzer, P. (2017). Interoception and gut feelings: Unconscious body sig‐ bial properties of picaridin and DEET against a broad range of mi‐
nals' impact on brain function, behavior and belief processes. In H.-F. croorganisms. World Journal of Microbiology & Biotechnology, 30(2),
Angel, L. Oviedo, R. F. Paloutzian, A. L. C. Runehov, & R. J. Seitz (Eds.), 407–411. https​://doi.org/10.1007/s11274-013-1456-4
New approaches to the scientific study of religion: Vol. 1. Processes of Kali, A. (2016). Psychobiotics: An emerging probiotic in psychiatric prac‐
believing: The acquisition, maintenance, and change in creditions (pp. tice. Biomedical Journal, 39(3), 223–224. https​://doi.org/10.1016/j.
435–442). Cham, Switzerland: Springer International Publishing. bj.2015.11.004
Holzer, P., & Farzi, A. (2014). Neuropeptides and the microbiota‐gut‐brain Kallivayalil, R. A. (2008). Are we over‐dependent on phar‐
axis. Advances in Experimental Medicine and Biology, 817, 195–219. macotherapy? Indian J Psychiatry, 50(1), 7–9. https​ ://doi.
https​://doi.org/10.1007/978-1-4939-0897-4_9 org/10.4103/0019-5545.39750​
Hood, S. D., Hince, D. A., Robinson, H., Cirillo, M., Christmas, D., & Karlsson, J., Sjostrom, L., & Sullivan, M. (1998). Swedish obese sub‐
Kaye, J. M. (2006). Serotonin regulation of the human stress re‐ jects (SOS)–an intervention study of obesity. Two‐year follow‐up of
sponse. Psychoneuroendocrinology, 31(9), 1087–1097. https​ ://doi. health‐related quality of life (HRQL) and eating behavior after gastric
org/10.1016/j.psyne​uen.2006.07.001 surgery for severe obesity. International Journal of Obesity and Related
Houghteling, P. D., & Walker, W. A. (2015). Why is initial bacterial colo‐ Metabolic Disorders, 22(2), 113–126.
nization of the intestine important to the infant's and child's health? Kelly, J. R., Clarke, G., Cryan, J. F., & Dinan, T. G. (2016). Brain‐gut‐mi‐
Journal of Pediatric Gastroenterology and Nutrition, 60(3), 294–307. crobiota axis: Challenges for translation in psychiatry. Annals of
https​://doi.org/10.1097/MPG.00000​0 0000​0 00597 Epidemiology, 26(5), 366–372. https​ ://doi.org/10.1016/j.annep​
Hyland, N. P., & Cryan, J. F. (2010). A gut feeling about GABA: Focus idem.2016.02.008
on GABA(B) receptors. Frontiers in Pharmacology, 1, 124. https​://doi. Khan, N. A., Raine, L. B., Drollette, E. S., Scudder, M. R., Kramer, A. F.,
org/10.3389/fphar.2010.00124​ & Hillman, C. H. (2015). Dietary fiber is positively associated with
Hyman, S. E. (2000). The genetics of mental illness: Implications for prac‐ cognitive control among prepubertal children. Journal of Nutrition,
tice. Bulletin of the World Health Organization, 78(4), 455–463. 145(1), 143–149. https​://doi.org/10.3945/jn.114.198457
Jacka, F. N., Cherbuin, N., Anstey, K. J., Sachdev, P., & Butterworth, P. Kim, H., Yun, Y., Ryu, S., Chang, Y., Kwon, M., Cho, J., … Kim, H. (2018).
(2015). Western diet is associated with a smaller hippocampus: Correlation between gut microbiota and personality in adults: A
A longitudinal investigation. BMC Medicine, 13, 215. https​ ://doi. cross‐sectional study. Brain, Behavior, and Immunity, 69, 374–385.
org/10.1186/s12916-015-0461-x https​://doi.org/10.1016/j.bbi.2017.12.012
Jacka, F. N., Kremer, P. J., Berk, M., de Silva‐Sanigorski, A. M., Moodie, Kleiman, S. C., Watson, H. J., Bulik‐Sullivan, E. C., Huh, E. Y., Tarantino,
M., Leslie, E. R., … Swinburn, B. A. (2011). A prospective study of diet L. M., Bulik, C. M., & Carroll, I. M. (2015). The intestinal microbi‐
quality and mental health in adolescents. PLoS ONE, 6(9), e24805. ota in acute anorexia nervosa and during renourishment: relation‐
https​://doi.org/10.1371/journ​al.pone.0024805 ship to depression, anxiety, and eating disorder psychopathology.
Jacka, F. N., Mykletun, A., Berk, M., Bjelland, I., & Tell, G. S. (2011). The as‐ Psychosomatic Medicine, 77(9), 969–981. https​ ://doi.org/10.1097/
sociation between habitual diet quality and the common mental dis‐ PSY.00000​0 0000​0 00247
orders in community‐dwelling adults: The Hordaland Health study. Knowles, S. R., Nelson, E. A., & Palombo, E. A. (2008). Investigating the
Psychosomatic Medicine, 73(6), 483–490. https​ ://doi.org/10.1097/ role of perceived stress on bacterial flora activity and salivary corti‐
PSY.0b013​e3182​22831a sol secretion: A possible mechanism underlying susceptibility to ill‐
Jacka, F. N., O'Neil, A., Opie, R., Itsiopoulos, C., Cotton, S., Mohebbi, ness. Biological Psychology, 77(2), 132–137. https​://doi.org/10.1016/j.
M., … Berk, M. (2017). A randomised controlled trial of dietary im‐ biops​ycho.2007.09.010
provement for adults with major depression (the ‘SMILES’ trial). BMC Kolokotroni, O., Middleton, N., Gavatha, M., Lamnisos, D., Priftis, K. N.,
Medicine, 15(1), 23. https​://doi.org/10.1186/s12916-017-0791-y & Yiallouros, P. K. (2012). Asthma and atopy in children born by cae‐
James, G. D. (2013). Ambulatory blood pressure variation: Allostasis sarean section: Effect modification by family history of allergies – a
and adaptation. Autonomic Neuroscience, 177(2), 87–94. https​://doi. population based cross‐sectional study. BMC Pediatrics, 12, 179–179.
org/10.1016/j.autneu.2013.03.012 https​://doi.org/10.1186/1471-2431-12-179
Jangi, S., Gandhi, R., Cox, L. M., Li, N., von Glehn, F., Yan, R., … Weiner, Kondo, T., Kishi, M., Fushimi, T., Ugajin, S., & Kaga, T. (2009). Vinegar
H. L. (2016). Alterations of the human gut microbiome in multiple intake reduces body weight, body fat mass, and serum triglyceride
sclerosis. Nature Communications, 7, 12015. https​://doi.org/10.1038/ levels in obese Japanese subjects. Bioscience, Biotechnology, and
ncomm​s12015 Biochemistry, 73(8), 1837–1843. https​://doi.org/10.1271/bbb.90231​
16 of 19 | GANCI et al.

Koopman, M., & El Aidy, S. (2017). Depressed gut? The microbiota‐diet‐ Lloyd‐Price, J., Abu‐Ali, G., & Huttenhower, C. (2016). The healthy human
inflammation trialogue in depression. Current Opinion in Psychiatry, microbiome. Genome Medicine, 8(1), 51. https​ ://doi.org/10.1186/
30(5), 369–377. https​://doi.org/10.1097/yco.00000​0 0000​0 00350 s13073-016-0307-y
Krishnan, S., Alden, N., & Lee, K. (2015). Pathways and functions of Luczynski, P., McVey Neufeld, K. A., Oriach, C. S., Clarke, G., Dinan, T.
gut microbiota metabolism impacting host physiology. Current G., & Cryan, J. F. (2016). Growing up in a bubble: Using germ‐free
Opinion in Biotechnology, 36, 137–145. https​ ://doi.org/10.1016/j. animals to assess the influence of the gut microbiota on brain and
copbio.2015.08.015 behavior. International Journal of Neuropsychopharmacology, 19(8),
Kuhnke, D., Jedlitschky, G., Grube, M., Krohn, M., Jucker, M., Mosyagin, pyw020. https​://doi.org/10.1093/ijnp/pyw020
I., … Vogelgesang, S. (2007). MDR1‐P‐glycoprotein (ABCB1) mediates Lyte, M. (2011). Probiotics function mechanistically as delivery vehi‐
transport of Alzheimer's Amyloid‐β peptides—implications for the cles for neuroactive compounds: Microbial endocrinology in the
mechanisms of Aβ clearance at the blood‐brain barrier. Brain Pathology, design and use of probiotics. BioEssays, 33(8), 574–581. https​://doi.
17(4), 347–353. https​://doi.org/10.1111/j.1750-3639.2007.00075.x org/10.1002/bies.20110 ​0 024
Lai, J. S., Hiles, S., Bisquera, A., Hure, A. J., McEvoy, M., & Attia, J. (2014). Macedo, D., Filho, A. J., Soares de Sousa, C. N., Quevedo, J., Barichello,
A systematic review and meta‐analysis of dietary patterns and de‐ T., Junior, H. V., & Freitas de Lucena, D. (2017). Antidepressants, anti‐
pression in community‐dwelling adults. American Journal of Clinical microbials or both? Gut microbiota dysbiosis in depression and possi‐
Nutrition, 99(1), 181–197. https​://doi.org/10.3945/ajcn.113.069880 ble implications of the antimicrobial effects of antidepressant drugs
Lam, Y. Y., Maguire, S., Palacios, T., & Caterson, I. D. (2017). Are the gut for antidepressant effectiveness. Journal of Affective Disorders, 208,
bacteria telling us to eat or not to eat? Reviewing the role of gut mi‐ 22–32. https​://doi.org/10.1016/j.jad.2016.09.012
crobiota in the etiology, disease progression and treatment of eating Maes, M., Kubera, M., Leunis, J. C., Berk, M., Geffard, M., & Bosmans,
disorders. Nutrients, 9(6), 602. https​://doi.org/10.3390/nu906​0602 E. (2013). In depression, bacterial translocation may drive inflam‐
Larsen, N., Vogensen, F. K., van den Berg, F. W. J., Nielsen, D. S., matory responses, oxidative and nitrosative stress (O&NS), and
Andreasen, A. S., Pedersen, B. K., … Jakobsen, M. (2010). Gut mi‐ autoimmune responses directed against O&NS‐damaged neoepi‐
crobiota in human adults with type 2 diabetes differs from non‐di‐ topes. Acta Psychiatrica Scandinavica, 127(5), 344–354. https​://doi.
abetic adults. PLoS ONE, 5(2), e9085. https​://doi.org/10.1371/journ​ org/10.1111/j.1600-0447.2012.01908.x
al.pone.0009085 Marks, D. M., Shah, M. J., Patkar, A. A., Masand, P. S., Park, G. Y., & Pae,
Lee, J., Duan, W., & Mattson, M. P. (2002). Evidence that brain‐derived C. U. (2009). Serotonin‐norepinephrine reuptake inhibitors for pain
neurotrophic factor is required for basal neurogenesis and mediates, control: Premise and promise. Current Neuropharmacology, 7(4), 331–
in part, the enhancement of neurogenesis by dietary restriction in 336. https​://doi.org/10.2174/15701​59097​90031201
the hippocampus of adult mice. Journal of Neurochemistry, 82(6), Markus, C. R., Panhuysen, G., Tuiten, A., Koppeschaar, H., Fekkes, D., &
1367–1375. https​://doi.org/10.1046/j.1471-4159.2002.01085.x Peters, M. L. (1998). Does carbohydrate‐rich, protein‐poor food pre‐
Lee, Y.‐T., Hu, L.‐Y., Shen, C.‐C., Huang, M.‐W., Tsai, S.‐J., Yang, A. C., vent a deterioration of mood and cognitive performance of stress‐
… Hung, J.‐H. (2015). Risk of psychiatric disorders following ir‐ prone subjects when subjected to a stressful task? Appetite, 31(1),
ritable bowel syndrome: A nationwide population‐based cohort 49–65. https​://doi.org/10.1006/appe.1997.0155
study. PLoS ONE, 10(7), e0133283. https​ ://doi.org/10.1371/journ​ Marques, T. M., Cryan, J. F., Shanahan, F., Fitzgerald, G. F., Ross, R. P.,
al.pone.0133283 Dinan, T. G., & Stanton, C. (2014). Gut microbiota modulation and
Leitão‐Gonçalves, R., Carvalho‐Santos, Z., Francisco, A. P., Fioreze, G. T., implications for host health: Dietary strategies to influence the gut–
Anjos, M., Baltazar, C., … Ribeiro, C. (2017). Commensal bacteria and brain axis. Innovative Food Science & Emerging Technologies, 22, 239–
essential amino acids control food choice behavior and reproduc‐ 247. https​://doi.org/10.1016/j.ifset.2013.10.016
tion. PLoS Biology, 15(4), e2000862. https​://doi.org/10.1371/journ​ Marvasti, F. F., & Stafford, R. S. (2012). From sick care to health care — re‐
al.pbio.2000862 engineering prevention into the U.S. system. New England Journal of
Leonard, B. E. (2010). The concept of depression as a dysfunction of the Medicine, 367(10), 889–891. https​://doi.org/10.1056/NEJMp​1206230
immune system. Current Immunology Reviews, 6(3), 205–212. https​:// Mathieson, F., Collings, S., & Dowell, A. (2009). Sub‐threshold mental
doi.org/10.2174/15733​95107​91823835 health syndromes: Finding an alternative to the medication of un‐
Ley, R. E., Turnbaugh, P. J., Klein, S., & Gordon, J. I. (2006). Microbial happiness. Journal of Primary Health Care, 1(1), 74–77.
ecology: Human gut microbes associated with obesity. Nature, Mayer, E. A. (2011). Gut feelings: The emerging biology of gut‐brain com‐
444(7122), 1022–1023. https​://doi.org/10.1038/4441022a munication. Nature Reviews Neuroscience, 12(8), 453–466. https​://
Lieberman, J. A., Stroup, T. S., McEvoy, J. P., Swartz, M. S., Rosenheck, doi.org/10.1038/nrn3071
R. A., Perkins, D. O., … Hsiao, J. K. (2005). Effectiveness of antipsy‐ Mayer, E. A., Naliboff, B. D., & Craig, A. D. (2006). Neuroimaging of
chotic drugs in patients with chronic schizophrenia. New England the brain‐gut axis: From basic understanding to treatment of func‐
Journal of Medicine, 353(12), 1209–1223. https​://doi.org/10.1056/ tional GI disorders. Gastroenterology, 131(6), 1925–1942. https​://doi.
NEJMo​a 051688 org/10.1053/j.gastro.2006.10.026
Linneberg, A., Nielsen, N. H., Madsen, F., Frølund, L., Dirksen, A., & Mayer, E. A., & Tillisch, K. (2011). The brain‐gut axis in abdominal pain
Jørgensen, T. (2000). Increasing prevalence of specific IgE to aeroal‐ syndromes. Annual Review of Medicine, 62, 381–396. https​ ://doi.
lergens in an adult population: Two cross‐sectional surveys 8 years org/10.1146/annur​ev-med-012309-103958
apart: The Copenhagen Allergy Study. The Journal of Allergy and Mazzoli, R., & Pessione, E. (2016). The neuro‐endocrinological role of
Clinical Immunology, 106(2), 247–252. https​ ://doi.org/10.1067/ microbial glutamate and GABA signaling. Frontiers in Microbiology, 7,
mai.2000.108312 1934. https​://doi.org/10.3389/fmicb.2016.01934​
Liou, A. P., Paziuk, M., Luevano, J.‐M., Machineni, S., Turnbaugh, P. J., & McEwen, B. S. (1998). Stress, adaptation, and disease. Allostasis and al‐
Kaplan, L. M. (2013). Conserved shifts in the gut microbiota due to lostatic load. Annals of the New York Academy of Sciences, 840, 33–44.
gastric bypass reduce host weight and adiposity. Science Translational https​://doi.org/10.1111/j.1749-6632.1998.tb095​46.x
Medicine, 5(178), 178ra141. https​://doi.org/10.1126/scitr​anslm​ McEwen, B. S. (2005). Stressed or stressed out: What is the difference?
ed.3005687 Journal of Psychiatry and Neuroscience, 30(5), 315–318.
Liu, Y. Z., Wang, Y. X., & Jiang, C. L. (2017). Inflammation: The common McEwen, B. S., & Wingfield, J. C. (2003). The concept of allostasis in bi‐
pathway of stress‐related diseases. Frontiers in Human Neuroscience, ology and biomedicine. Hormones and Behavior, 43(1), 2–15. https​://
11, 316. https​://doi.org/10.3389/fnhum.2017.00316​ doi.org/10.1016/S0018-506X(02)00024-7
GANCI et al. | 17 of 19

McLeod, J. D., & Fettes, D. L. (2007). Trajectories of failure: The educational volunteers. International Journal of Neuropsychopharmacology, 12(2),
careers of children with mental health problems. American Journal of 169–179. https​://doi.org/10.1017/S1461​14570​8 009164
Sociology, 113(3), 653–701. https​://doi.org/10.1086/521849 Myles, I. A. (2014). Fast food fever: Reviewing the impacts of the
Meldrum, B. S. (2000). Glutamate as a neurotransmitter in the brain: Western diet on immunity. Nutrition Journal, 13, 61. https​ ://doi.
Review of physiology and pathology. Journal of Nutrition, 130(4S org/10.1186/1475-2891-13-61
Suppl), 1007s–1015s. https​://doi.org/10.1093/jn/130.4.1007S​ Neufeld, K. M., Kang, N., Bienenstock, J., & Foster, J. A. (2011). Reduced
Mendlowicz, M. V., & Stein, M. B. (2000). Quality of life in individuals anxiety‐like behavior and central neurochemical change in germ‐free
with anxiety disorders. The American Journal of Psychiatry, 157(5), mice. Neurogastroenterology and Motility, 23(3), 255–264, e119. https​
669–682. https​://doi.org/10.1176/appi.ajp.157.5.669 ://doi.org/10.1111/j.1365-2982.2010.01620.x
Meneses, A., & Liy‐Salmeron, G. (2012). Serotonin and emotion, learning Niccoli, T., & Partridge, L. (2012). Ageing as a risk factor for disease.
and memory. Reviews in the Neurosciences, 23(5–6), 543–553. https​:// Current Biology, 22(17), R741–752. https​ ://doi.org/10.1016/j.
doi.org/10.1515/revne​uro-2012-0060 cub.2012.07.024
Messaoudi, M., Violle, N., Bisson, J. F., Desor, D., Javelot, H., & Rougeot, Noakes, T. D. (2012). Fatigue is a brain‐derived emotion that regulates
C. (2011). Beneficial psychological effects of a probiotic formulation the exercise behavior to ensure the protection of whole body ho‐
(Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in meostasis. Frontiers in Physiology, 3, 82. https​ ://doi.org/10.3389/
healthy human volunteers. Gut Microbes, 2(4), 256–261. https​://doi. fphys.2012.00082​
org/10.4161/gmic.2.4.16108​ Ochner, C. N., Kwok, Y., Conceição, E., Pantazatos, S. P., Puma, L. M.,
Meyer, U., & Feldon, J. (2009). Prenatal exposure to infection: A primary Carnell, S., … Geliebter, A. (2011). Selective reduction in neural re‐
mechanism for abnormal dopaminergic development in schizo‐ sponses to high calorie foods following gastric bypass surgery. Annals
phrenia. Psychopharmacology (Berl), 206(4), 587–602. https​ ://doi. of Surgery, 253(3), 502–507. https​ ://doi.org/10.1097/SLA.0b013​
org/10.1007/s00213-009-1504-9 e3182​03a289
Mika, A., Van Treuren, W., Gonzalez, A., Herrera, J. J., Knight, R., & Ochner, C. N., Stice, E., Hutchins, E., Afifi, L., Geliebter, A., Hirsch, J.,
Fleshner, M. (2015). Exercise is more effective at altering gut micro‐ & Teixeira, J. (2012). Relation between changes in neural respon‐
bial composition and producing stable changes in lean mass in juve‐ sivity and reductions in desire to eat high‐calorie foods following
nile versus adult male f344 rats. PLoS ONE, 10(5), e0125889. https​:// gastric bypass surgery. Neuroscience, 209, 128–135. https​ ://doi.
doi.org/10.1371/journ​al.pone.0125889 org/10.1016/j.neuro​scien​ce.2012.02.030
Miller, A. H., & Raison, C. L. (2016). The role of inflammation in depres‐ OECD (2017). Health at a glance 2017: OECD indicators. Paris, France:
sion: From evolutionary imperative to modern treatment target. OECD Publishing.
Nature Reviews Immunology, 16(1), 22–34. https​://doi.org/10.1038/ O'Mahony, S. M., Clarke, G., Borre, Y. E., Dinan, T. G., & Cryan, J. F.
nri.2015.5 (2015). Serotonin, tryptophan metabolism and the brain‐gut‐mi‐
Mokhber, N., Shaghayegh, H., Talebi, M., & Tavassoli, A. (2016). crobiome axis. Behavioral Brain Research, 277, 32–48. https​ ://doi.
Comparison of levels of depression in patients with excessive obe‐ org/10.1016/j.bbr.2014.07.027
sity before and after gastric bypass surgery. Journal of Minimally Oriach, C. S., Robertson, R. C., Stanton, C., Cryan, J. F., & Dinan, T. G.
Invasive Surgical Sciences, 5(4), e34947. https​ ://doi.org/10.17795/​ (2016). Food for thought: The role of nutrition in the microbiota‐
minsu​rgery-34947​ gut–brain axis. Clinical Nutrition Experimental, 6, 25–38. https​://doi.
Moloney, R. D., Desbonnet, L., Clarke, G., Dinan, T. G., & Cryan, J. F. (2014). org/10.1016/j.yclnex.2016.01.003
The microbiome: Stress, health and disease. Mammalian Genome, Ortiz, G. G., Benítez‐King, G. A., Rosales‐Corral, S. A., Pacheco‐Moisés,
25(1–2), 49–74. https​://doi.org/10.1007/s00335-013-9488-5 F. P., & Velázquez‐Brizuela, I. E. (2008). Cellular and biochemical ac‐
Monda, V., Villano, I., Messina, A., Valenzano, A., Esposito, T., Moscatelli, tions of melatonin which protect against free radicals: role in neuro‐
F., … Messina, G. (2017). Exercise modifies the gut microbiota with degenerative disorders. Current Neuropharmacology, 6(3), 203–214.
positive health effects. Oxidative Medicine and Cellular Longevity, https​://doi.org/10.2174/15701​59087​85777201
2017, 8. https​://doi.org/10.1155/2017/3831972 Palmeira, A. L., Branco, T. L., Martins, S. C., Minderico, C. S., Silva, M.
Moret, C., & Briley, M. (2011). The importance of norepinephrine in de‐ N., Vieira, P. N., … Teixeira, P. J. (2010). Change in body image and
pression. Neuropsychiatric Disease and Treatment, 7(Suppl 1), 9–13. psychological well‐being during behavioral obesity treatment:
https​://doi.org/10.2147/NDT.S19619 Associations with weight loss and maintenance. Body Image, 7(3),
Morrison, D. J., & Preston, T. (2016). Formation of short chain fatty 187–193. https​://doi.org/10.1016/j.bodyim.2010.03.002
acids by the gut microbiota and their impact on human metabo‐ Palsson, O. S., & Whitehead, W. E. (2013). Psychological treatments in
lism. Gut Microbes, 7(3), 189–200. https​ ://doi.org/10.1080/19490​ functional gastrointestinal disorders: A primer for the gastroenterol‐
976.2015.1134082 ogist. Clinical Gastroenterology and Hepatology, 11(3), 208–216; quiz
Mu, C., Yang, Y., & Zhu, W. (2016). Gut microbiota: the brain peace‐ e222–203. https​://doi.org/10.1016/j.cgh.2012.10.031
keeper. Frontiers in Microbiology, 7, 345. https​ ://doi.org/10.3389/ Patel, J. P., & Frey, B. N. (2015). Disruption in the blood‐brain barrier: The
fmicb.2016.00345​ missing link between brain and body inflammation in bipolar disorder?
Mueller, N. T., Bakacs, E., Combellick, J., Grigoryan, Z., & Dominguez‐ Neural Plasticity, 2015, 12. https​://doi.org/10.1155/2015/708306
Bello, M. G. (2015). The infant microbiome development: Mom Paulus, M. P., & Stein, M. B. (2010). Interoception in anxiety and depres‐
matters. Trends in Molecular Medicine, 21(2), 109–117. https​://doi. sion. Brain Structure and Function, 214(5–6), 451–463. https​://doi.
org/10.1016/j.molmed.2014.12.002 org/10.1007/s00429-010-0258-9
Mulak, A., & Bonaz, B. (2015). Brain‐gut‐microbiota axis in Parkinson's Peng, L., Li, Z. R., Green, R. S., Holzman, I. R., & Lin, J. (2009). Butyrate
disease. World Journal of Gastroenterology, 21(37), 10609–10620. enhances the intestinal barrier by facilitating tight junction assem‐
https​://doi.org/10.3748/wjg.v21.i37.10609​ bly via activation of AMP‐activated protein kinase in Caco‐2 cell
Müller, C. P., & Homberg, J. R. (2015). The role of serotonin in drug use monolayers. Journal of Nutrition, 139(9), 1619–1625. https​ ://doi.
and addiction. Behavioral Brain Research, 277, 146–192. https​://doi. org/10.3945/jn.109.104638
org/10.1016/j.bbr.2014.04.007 Polidano, C., Zhu, A., & Bornstein, J. C. (2017). The relation be‐
Murphy, S. E., Yiend, J., Lester, K. J., Cowen, P. J., & Harmer, C. J. (2009). tween caesarean birth and child cognitive development. Scientific
Short‐term serotonergic but not noradrenergic antidepressant Reports, 7(11483), 1–10. https​ ://doi.org/10.1038/s41598-017-
administration reduces attentional vigilance to threat in healthy 10831-y
18 of 19 | GANCI et al.

Poti, J. M., Mendez, M. A., Ng, S. W., & Popkin, B. M. (2015). Is the de‐ Shahar, D., Shai, I., Vardi, H., Shahar, A., & Fraser, D. (2005). Diet and
gree of food processing and convenience linked with the nutritional eating habits in high and low socioeconomic groups. Nutrition, 21(5),
quality of foods purchased by US households? American Journal 559–566. https​://doi.org/10.1016/j.nut.2004.09.018
of Clinical Nutrition, 101(6), 1251–1262. https​ ://doi.org/10.3945/ Sheedy, J. R., Wettenhall, R. E., Scanlon, D., Gooley, P. R., Lewis, D. P.,
ajcn.114.100925 McGregor, N., … De Meirleir, K. L. (2009). Increased d‐lactic acid in‐
Quercia, S., Candela, M., Giuliani, C., Turroni, S., Luiselli, D., Rampelli, testinal bacteria in patients with chronic fatigue syndrome. In Vivo,
S., … Pirazzini, C. (2014). From lifetime to evolution: Timescales of 23(4), 621–628.
human gut microbiota adaptation. Frontiers in Microbiology, 5, 1–9. Sheflin, A., Melby, C. L., Carbonero, F., & Weir, T. L. (2017). Linking dietary
https​://doi.org/10.3389/fmicb.2014.00587​ patterns with gut microbial composition and function. Gut Microbes,
Rao, A. V., Bested, A. C., Beaulne, T. M., Katzman, M. A., Iorio, C., 8(2), 113–129. https​://doi.org/10.1080/19490​976.2016.1270809
Berardi, J. M., & Logan, A. C. (2009). A randomized, double‐blind, Shi, Y., Dong, Y., Huang, W., Zhu, D., Mao, H., & Su, P. (2016). Fecal micro‐
placebo‐controlled pilot study of a probiotic in emotional symp‐ biota transplantation for ulcerative colitis: A systematic review and
toms of chronic fatigue syndrome. Gut Pathogens, 1(1), 6. https​://doi. meta‐analysis. PLoS ONE, 11(6), e0157259. https​://doi.org/10.1371/
org/10.1186/1757-4749-1-6 journ​al.pone.0157259
Rea, K., Dinan, T. G., & Cryan, J. F. (2016). The microbiome: A key regula‐ Shishov, V. A., Kirovskaya, T. A., Kudrin, V. S., & Oleskin, A. V. (2009).
tor of stress and neuroinflammation. Neurobiol Stress, 4, 23–33. https​ Amine neuromediators, their precursors, and oxidation products
://doi.org/10.1016/j.ynstr.2016.03.001 in the culture of Escherichia coli K‐12. Applied Biochemistry and
Rhee, S. H., Pothoulakis, C., & Mayer, E. A. (2009). Principles and clin‐ Microbiology, 45(5), 494–497. https​://doi.org/10.1134/S0003​68380​
ical implications of the brain‐gut‐enteric microbiota axis. Nature 9050068
Reviews Gastroenterology & Hepatology, 6(5), 306–314. https​://doi. Singh, A. R. (2010). Modern medicine: Towards prevention, cure, well‐
org/10.1038/nrgas​tro.2009.35 being and longevity. Mens Sana Monographs, 8(1), 17–29. https​://doi.
Rogers, G. B., Keating, D. J., Young, R. L., Wong, M. L., Licinio, J., & org/10.4103/0973-1229.58817​
Wesselingh, S. (2016). From gut dysbiosis to altered brain function Singh, R. K., Chang, H.‐W., Yan, D. I., Lee, K. M., Ucmak, D., Wong, K., …
and mental illness: Mechanisms and pathways. Molecular Psychiatry, Liao, W. (2017). Influence of diet on the gut microbiome and impli‐
21(6), 738–748. https​://doi.org/10.1038/mp.2016.50 cations for human health. Journal of Translational Medicine, 15(1), 73.
Roohafza, H., Bidaki, E. Z., Hasanzadeh‐Keshteli, A., Daghaghzade, H., https​://doi.org/10.1186/s12967-017-1175-y
Afshar, H., & Adibi, P. (2016). Anxiety, depression and distress among Sirin, S. R. (2005). Socioeconomic status and academic achievement: a
irritable bowel syndrome and their subtypes: An epidemiological meta‐analytic review of research. Review of Educational Research,
population based study. Advanced Biomedical Research, 5, 183. https​ 75(3), 417–453. https​://doi.org/10.3102/00346​54307​5003417
://doi.org/10.4103/2277-9175.190938 Smith, P. M., Howitt, M. R., Panikov, N., Michaud, M., Gallini, C. A.,
Rubio‐Ruiz, M. E., Peredo‐Escárcega, A. E., Cano‐Martínez, A., & Bohlooly‐Y, M., … Garrett, W. S. (2013). The microbial metabo‐
Guarner‐Lans, V. (2015). An evolutionary perspective of nutri‐ lites, short‐chain fatty acids, regulate colonic Treg cell homeosta‐
tion and inflammation as mechanisms of cardiovascular disease. sis. Science, 341(6145), 569–573. https​ ://doi.org/10.1126/scien​
International Journal of Evolutionary Biology, 2015, 1–10. https​://doi. ce.1241165
org/10.1155/2015/179791 Song, P., Patel, N. B., Gunther, S., Li, C.‐S., Liu, Y., Lee, C. Y. G., … Wong,
Sampson, T. R., & Mazmanian, S. K. (2015). Control of brain development, M. S. (2016). Body image and quality of life: changes with gastric
function, and behavior by the microbiome. Cell Host & Microbe, 17(5), bypass and body contouring. Annals of Plastic Surgery, 76(Suppl 3),
565–576. https​://doi.org/10.1016/j.chom.2015.04.011 S216–S221. https​://doi.org/10.1097/SAP.00000​0 0000​0 00788
Sarwer, D. B., & Steffen, K. J. (2015). Quality of life, body image and Song, S. J., Lauber, C., Costello, E. K., Lozupone, C. A., Humphrey, G.,
sexual functioning in bariatric surgery patients. European Eating Berg‐Lyons, D., … Knight, R. (2013). Cohabiting family members
Disorders Review, 23(6), 504–508. https​://doi.org/10.1002/erv.2412 share microbiota with one another and with their dogs. eLife, 2,
Scheperjans, F., Aho, V., Pereira, P. A. B., Koskinen, K., Paulin, L., e00458. https​://doi.org/10.7554/eLife.00458​.
Pekkonen, E., … Auvinen, P. (2015). Gut microbiota are related to Srivastava, K., & Das, R. C. (2015). Personality and health: Road to
Parkinson's disease and clinical phenotype. Movement Disorders, well‐being. Industrial Psychiatry Journal, 24(1), 1–4. https​ ://doi.
30(3), 350–358. https​://doi.org/10.1002/mds.26069​ org/10.4103/0972-6748.160905
Schmidt, K., Cowen, P. J., Harmer, C. J., Tzortzis, G., Errington, S., & Stanislawski, M. A., Dabelea, D., Wagner, B. D., Sontag, M. K., Lozupone,
Burnet, P. W. (2015). Prebiotic intake reduces the waking cor‐ C. A., & Eggesbø, M. (2017). Pre‐pregnancy weight, gestational
tisol response and alters emotional bias in healthy volunteers. weight gain, and the gut microbiota of mothers and their infants.
Psychopharmacology (Berl), 232(10), 1793–1801. https​ ://doi. Microbiome, 5(1), 113. https​://doi.org/10.1186/s40168-017-0332-0
org/10.1007/s00213-014-3810-0 Stasi, C., Rosselli, M., Zignego, A. L., Laffi, G., & Milani, S. (2014). Serotonin
Schnorr, S. L., Sankaranarayanan, K., Lewis, C. M. Jr, & Warinner, C. and its implication in the side‐effects of interferon‐based treatment
(2016). Insights into human evolution from ancient and contempo‐ of patients with chronic viral hepatitis: Pharmacological interven‐
rary microbiome studies. Current Opinion in Genetics & Development, tions. Hepatology Research, 44(1), 9–16. https​ ://doi.org/10.1111/
41, 14–26. https​://doi.org/10.1016/j.gde.2016.07.003 hepr.12116​
Schoknecht, K., & Shalev, H. (2012). Blood‐brain barrier dysfunction in Steenbergen, L., Sellaro, R., van Hemert, S., Bosch, J. A., & Colzato, L. S.
brain diseases: Clinical experience. Epilepsia, 53(Suppl 6), 7–13. https​ (2015). A randomized controlled trial to test the effect of multispecies
://doi.org/10.1111/j.1528-1167.2012.03697.x probiotics on cognitive reactivity to sad mood. Brain, Behavior, and
Sekirov, I., Russell, S. L., Antunes, L. C., & Finlay, B. B. (2010). Gut mi‐ Immunity, 48, 258–264. https​://doi.org/10.1016/j.bbi.2015.04.003
crobiota in health and disease. Physiological Reviews, 90(3), 859–904. Sudo, N. (2012). Role of microbiome in regulating the HPA axis and its
https​://doi.org/10.1152/physr​ev.00045.2009 relevance to allergy. Chemical Immunology and Allergy, 98, 163–175.
Sender, R., Fuchs, S., & Milo, R. (2016). Revised estimates for the num‐ https​://doi.org/10.1159/00033​6510
ber of human and bacteria cells in the body. PLoS Biology, 14(8), Sudo, N., Chida, Y., Aiba, Y., Sonoda, J., Oyama, N., Yu, X.‐N., … Koga,
e1002533. https​://doi.org/10.1371/journ​al.pbio.1002533 Y. (2004). Postnatal microbial colonization programs the hypotha‐
Sevelsted, A., Stokholm, J., Bonnelykke, K., & Bisgaard, H. (2015). lamic‐pituitary‐adrenal system for stress response in mice. Journal
Cesarean section and chronic immune disorders. Pediatrics, 135(1), of Physiology, 558(Pt 1), 263–275. https​ ://doi.org/10.1113/jphys​
e92–e98. https​://doi.org/10.1542/peds.2014-0596 iol.2004.063388
GANCI et al. | 19 of 19

Sun, J., Wang, F., Li, H., Zhang, H., Jin, J., Chen, W., … Liu, C. (2015). Whitaker‐Azmitia, P. M. (2001). Serotonin and brain development: Role
Neuroprotective effect of sodium butyrate against cerebral isch‐ in human developmental diseases. Brain Research Bulletin, 56(5), 479–
emia/reperfusion injury in mice. BioMed Research International, 2015, 485. https​://doi.org/10.1016/S0361-9230(01)00615-3
395895. https​://doi.org/10.1155/2015/395895 Wong, R. K., Yang, C., Song, G.‐H., Wong, J., & Ho, K.‐Y. (2015).
Tamburini, S., Shen, N., Wu, H. C., & Clemente, J. C. (2016). The microbi‐ Melatonin regulation as a possible mechanism for probiotic (VSL#3)
ome in early life: Implications for health outcomes. Nature Medicine, in irritable bowel syndrome: a randomized double‐blinded placebo
22(7), 713–722. https​://doi.org/10.1038/nm.4142 study. Digestive Diseases and Sciences, 60(1), 186–194. https​://doi.
Tana, C., Umesaki, Y., Imaoka, A., Handa, T., Kanazawa, M., & Fukudo, org/10.1007/s10620-014-3299-8
S. (2010). Altered profiles of intestinal microbiota and organic World Health Organisation (WHO) (2017). Depression (Fact sheet No.
acids may be the origin of symptoms in irritable bowel syndrome. 369). Retrieved from http://www.who.int/media​centr​e/facts​heets/​
Neurogastroenterology and Motility, 22(5), 512–519, e114–515. https​ fs369/​en/
://doi.org/10.1111/j.1365-2982.2009.01427.x Wu, G. D., Chen, J., Hoffmann, C., Bittinger, K., Chen, Y.‐Y., Keilbaugh,
Theis, K. R., Dheilly, N. M., Klassen, J. L., Brucker, R. M., Baines, J. F., S. A., … Lewis, J. D. (2011). Linking long‐term dietary patterns with
Bosch, T. C. G., … Bordenstein, S. R. (2016). Getting the hologenome gut microbial enterotypes. Science, 334(6052), 105–108. https​://doi.
concept right: An eco‐evolutionary framework for hosts and their org/10.1126/scien​ce.1208344
microbiomes. mSystems, 1(2), e00028‐16.https​://doi.org/10.1128/ Yamamoto, K., & Hornykiewicz, O. (2004). Proposal for a noradrenaline
mSyst​ems.00028-16 hypothesis of schizophrenia. Progress in Neuro‐Psychopharmacology
Tillisch, K., Labus, J., Kilpatrick, L., Jiang, Z., Stains, J., Ebrat, B., … Mayer, and Biological Psychiatry, 28(5), 913–922. https​://doi.org/10.1016/j.
E. A. (2013). Consumption of fermented milk product with probiotic pnpbp.2004.05.033
modulates brain activity. Gastroenterology, 144(7), 1394–1401, 1401. Yamamura, S., Morishima, H., Kumano‐go, T., Suganuma, N., Matsumoto,
e1391–1394. https​://doi.org/10.1053/j.gastro.2013.02.043 H., Adachi, H., … Takeda, M. (2009). The effect of Lactobacillus helve‐
Ussar, S., Fujisaka, S., & Kahn, C. R. (2016). Interactions between host ticus fermented milk on sleep and health perception in elderly sub‐
genetics and gut microbiome in diabetes and metabolic syndrome. jects. European Journal of Clinical Nutrition, 63(1), 100–105. https​://
Molecular Metabolism, 5(9), 795–803. https​ ://doi.org/10.1016/j. doi.org/10.1038/sj.ejcn.1602898
molmet.2016.07.004 Yarandi, S. S., Peterson, D. A., Treisman, G. J., Moran, T. H., & Pasricha,
Uyeda, J. C., Hansen, T. F., Arnold, S. J., & Pienaar, J. (2011). The mil‐ P. J. (2016). Modulatory effects of gut microbiota on the central ner‐
lion‐year wait for macroevolutionary bursts. Proceedings of the vous system: How gut could play a role in neuropsychiatric health
National Academy of Sciences USA, 108(38), 15908–15913. https​:// and diseases. Journal of Neurogastroenterology and Motility, 22(2),
doi.org/10.1073/pnas.10145​03108​ 201–212. https​://doi.org/10.5056/jnm15146
Van Oudenhove, L., Crowell, M. D., Drossman, D. A., Halpert, A. D., Yatsunenko, T., Rey, F. E., Manary, M. J., Trehan, I., Dominguez‐Bello,
Keefer, L., Lackner, J. M., … Levy, R. L. (2016). Biopsychosocial as‐ M. G., Contreras, M., … Gordon, J. I. (2012). Human gut microbiome
pects of functional gastrointestinal disorders. Gastroenterology, viewed across age and geography. Nature, 486(7402), 222–227. https​
150(6), 1355–1367. https​://doi.org/10.1053/j.gastro.2016.02.027 ://doi.org/10.1038/natur​e11053
Vangay, P., Ward, T., Gerber, J. S., & Knights, D. (2015). Antibiotics, pe‐ Yoto, A., Murao, S., Motoki, M., Yokoyama, Y., Horie, N., Takeshima, K.,
diatric dysbiosis, and disease. Cell Host & Microbe, 17(5), 553–564. … Yokogoshi, H. (2012). Oral intake of gamma‐aminobutyric acid af‐
https​://doi.org/10.1016/j.chom.2015.04.006 fects mood and activities of central nervous system during stressed
Veatch, O. J., Goldman, S. E., Adkins, K. W., & Malow, B. A. (2015). condition induced by mental tasks. Amino Acids, 43(3), 1331–1337.
Melatonin in children with autism spectrum disorders: How does the https​://doi.org/10.1007/s00726-011-1206-6
evidence fit together? Journal of Nature and Science, 1(7), e125. Zagajewski, J., Drozdowicz, D., Brzozowska, I., Hubalewska‐Mazgaj, M.,
Voreades, N., Kozil, A., & Weir, T. L. (2014). Diet and the development Stelmaszynska, T., Laidler, P. M., & Brzozowski, T. (2012). Conversion
of the human intestinal microbiome. Frontiers in Microbiology, 5(494), L‐tryptophan to melatonin in the gastrointestinal tract: The new high
1–9. https​://doi.org/10.3389/fmicb.2014.00494​ performance liquid chromatography method enabling simultaneous
Vozeh, S. (2003). Is the increasing use of evidence‐based pharma‐ determination of six metabolites of L‐tryptophan by native fluores‐
cotherapy causing the renaissance of complementary medicine? cence and UV‐VIS detection. Journal of Physiology and Pharmacology,
British Journal of Clinical Pharmacology, 56(3), 292–296. https​://doi. 63(6), 613–621.
org/10.1046/j.0306-5251.2003.01879.x Zhang, H., DiBaise, J. K., Zuccolo, A., Kudrna, D., Braidotti, M., Yu, Y., …
Wallis, A., Butt, H., Ball, M., Lewis, D. P., & Bruck, D. (2016). Support Krajmalnik‐Brown, R. (2009). Human gut microbiota in obesity and
for the microgenderome: Associations in a human clinical population. after gastric bypass. Proceedings of the National Academy of Sciences
Scientific Reports, 6, 1–9. https​://doi.org/10.1038/srep1​9171 USA, 106(7), 2365–2370. https​://doi.org/10.1073/pnas.08126​0 0106​
Warren, R. P., & Singh, V. K. (1996). Elevated serotonin levels in au‐
tism: Association with the major histocompatibility complex.
Neuropsychobiology, 34(2), 72–75. https​ ://doi.org/10.1159/00011​
How to cite this article: Ganci M, Suleyman E, Butt H, Ball M.
9295
The role of the brain–gut–microbiota axis in psychology: The
Welly, R. J., Liu, T.‐W., Zidon, T. M., Rowles, J. L., Park, Y.‐M., Smith, T. N.,
… Vieira‐Potter, V. J. (2016). Comparison of diet vs. exercise on met‐ importance of considering gut microbiota in the development,
abolic function & gut microbiota in obese rats. Medicine and Science perpetuation, and treatment of psychological disorders. Brain
in Sports and Exercise, 48(9), 1688–1698. https​://doi.org/10.1249/ Behav. 2019;9:e01408. https​://doi.org/10.1002/brb3.1408
MSS.00000​0 0000​0 00964

You might also like